Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Biochemistry ; 55(11): 1645-51, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26813693

ABSTRACT

The protein methyltransferase (PMT) SETDB1 is a strong candidate oncogene in melanoma and lung carcinomas. SETDB1 methylates lysine 9 of histone 3 (H3K9), utilizing S-adenosylmethionine (SAM) as the methyl donor and its catalytic activity, has been reported to be regulated by a partner protein ATF7IP. Here, we examine the contribution of ATF7IP to the in vitro activity and substrate specificity of SETDB1. SETDB1 and ATF7IP were co-expressed and 1:1 stoichiometric complexes were purified for comparison against SETDB1 enzyme alone. We employed both radiometric flashplate-based and SAMDI mass spectrometry assays to follow methylation on histone H3 15-mer peptides, where lysine 9 was either unmodified, monomethylated, or dimethylated. Results show that SETDB1 and the SETDB1:ATF7IP complex efficiently catalyze both monomethylation and dimethylation of H3K9 peptide substrates. The activity of the binary complex was 4-fold lower than SETDB1 alone. This difference was due to a decrease in the value of kcat as the substrate KM values were comparable between SETDB1 and the SETDB1:ATF7IP complex. H3K9 methylation by SETDB1 occurred in a distributive manner, and this too was unaffected by the presence of ATF7IP. This finding is important as H3K9 can be methylated by HMTs other than SETDB1 and a distributive mechanism would allow for interplay between multiple HMTs on H3K9. Our results indicate that ATF7IP does not directly modulate SETDB1 catalytic activity, suggesting alternate roles, such as affecting cellular localization or mediating interaction with additional binding partners.


Subject(s)
Histones/chemistry , Multiprotein Complexes/chemistry , Protein Methyltransferases/chemistry , S-Adenosylmethionine/chemistry , Transcription Factors/chemistry , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Mass Spectrometry , Methylation , Multiprotein Complexes/metabolism , Protein Methyltransferases/metabolism , Repressor Proteins , S-Adenosylmethionine/metabolism , Substrate Specificity/physiology , Transcription Factors/metabolism
3.
J Control Release ; 220(Pt B): 758-65, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26385168

ABSTRACT

Protein methyltransferases are emerging as promising drug targets for therapeutic intervention in human cancers. Pinometostat (EPZ-5676) is a small molecule inhibitor of the DOT1L enzyme, a histone methyltransferase that methylates lysine 79 of histone H3. DOT1L activity is dysregulated in the pathophysiology of rearranged mixed lineage leukemia (MLL-r). Pinometostat is currently in Phase 1 clinical trials in relapsed refractory acute leukemia patients and is administered as a continuous IV infusion (CIV). The studies herein investigated alternatives to CIV administration of pinometostat to improve patient convenience. Various sustained release technologies were considered, and based on the required dose size as well as practical considerations, subcutaneous (SC) bolus administration of a solution formulation was selected for further evaluation in preclinical studies. SC administration offered improved exposure and complete bioavailability of pinometostat relative to CIV and oral administration. These findings warranted further evaluation in rat xenograft models of MLL-r leukemia. SC dosing in xenograft models demonstrated inhibition of MLL-r tumor growth and inhibition of pharmacodynamic markers of DOT1L activity. However, a dosing frequency of thrice daily (t.i.d) was required in these studies to elicit optimal inhibition of DOT1L target genes and tumor growth inhibition. Development of an extended release formulation may prove useful in the further optimization of the SC delivery of pinometostat, moving towards a more convenient dosing paradigm for patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzimidazoles/administration & dosage , DNA Methylation/drug effects , Drug Carriers , Enzyme Inhibitors/administration & dosage , Epigenesis, Genetic/drug effects , Leukemia, Biphenotypic, Acute/drug therapy , Methyltransferases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/blood , Benzimidazoles/chemistry , Benzimidazoles/pharmacokinetics , Biological Availability , Chemistry, Pharmaceutical , Delayed-Action Preparations , Dogs , Drug Administration Schedule , Enzyme Inhibitors/blood , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Gene Expression Regulation, Leukemic/drug effects , Histone-Lysine N-Methyltransferase , Humans , Infusions, Intravenous , Injections, Subcutaneous , Leukemia, Biphenotypic, Acute/enzymology , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Biphenotypic, Acute/pathology , Male , Methyltransferases/metabolism , Mice , Rats, Sprague-Dawley , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Biopharm Drug Dispos ; 35(4): 237-52, 2014 May.
Article in English | MEDLINE | ID: mdl-24415392

ABSTRACT

(2R,3R,4S,5R)-2-(6-Amino-9H-purin-9-yl)-5-((((1r,3S)-3-(2-(5-(tert-butyl)-1H-benzo[d]imidazol-2-yl)ethyl)cyclobutyl)(isopropyl)amino)methyl)tetrahydrofuran-3,4-diol (EPZ-5676) is a novel DOT1L histone methyltransferase inhibitor currently in clinical development for the treatment of MLL-rearranged leukemias. This report describes the preclinical pharmacokinetics and metabolism of EPZ-5676, an aminonucleoside analog with exquisite target potency and selectivity that has shown robust and durable tumor growth inhibition in preclinical models. The in vivo pharmacokinetics in mouse, rat and dog were characterized following i.v. and p.o. administration; EPZ-5676 had moderate to high clearance, low oral bioavailability with a steady-state volume of distribution 2-3 fold higher than total body water. EPZ-5676 showed biexponential kinetics following i.v. administration, giving rise to a terminal elimination half-life (t1/2 ) of 1.1, 3.7 and 13.6 h in mouse, rat and dog, respectively. The corresponding in vitro ADME parameters were also studied and utilized for in vitro-in vivo extrapolation purposes. There was good agreement between the microsomal clearance and the in vivo clearance implicating hepatic oxidative metabolism as the predominant elimination route in preclinical species. Furthermore, low renal clearance was observed in mouse, approximating to fu -corrected glomerular filtration rate (GFR) and thus passive glomerular filtration. The metabolic pathways across species were studied in liver microsomes in which EPZ-5676 was metabolized to three monohydroxylated metabolites (M1, M3 and M5), one N-dealkylated product (M4) as well as an N-oxide (M6).


Subject(s)
Antineoplastic Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Methyltransferases/antagonists & inhibitors , Animals , Antineoplastic Agents/blood , Benzimidazoles/blood , Blood Proteins/metabolism , Dogs , Hepatocytes/metabolism , Madin Darby Canine Kidney Cells , Male , Mice , Microsomes, Liver/metabolism , Permeability , Rats, Sprague-Dawley
5.
Blood ; 122(6): 1017-25, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23801631

ABSTRACT

Rearrangements of the MLL gene define a genetically distinct subset of acute leukemias with poor prognosis. Current treatment options are of limited effectiveness; thus, there is a pressing need for new therapies for this disease. Genetic and small molecule inhibitor studies have demonstrated that the histone methyltransferase DOT1L is required for the development and maintenance of MLL-rearranged leukemia in model systems. Here we describe the characterization of EPZ-5676, a potent and selective aminonucleoside inhibitor of DOT1L histone methyltransferase activity. The compound has an inhibition constant value of 80 pM, and demonstrates 37 000-fold selectivity over all other methyltransferases tested. In cellular studies, EPZ-5676 inhibited H3K79 methylation and MLL-fusion target gene expression and demonstrated potent cell killing that was selective for acute leukemia lines bearing MLL translocations. Continuous IV infusion of EPZ-5676 in a rat xenograft model of MLL-rearranged leukemia caused complete tumor regressions that were sustained well beyond the compound infusion period with no significant weight loss or signs of toxicity. EPZ-5676 is therefore a potential treatment of MLL-rearranged leukemia and is under clinical investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Leukemia/genetics , Leukemia/therapy , Methyltransferases/antagonists & inhibitors , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Dose-Response Relationship, Drug , Female , Histone Methyltransferases , Histones/metabolism , Humans , Neoplasm Transplantation , Protein Conformation , Rats , Rats, Nude
6.
Bioorg Med Chem Lett ; 23(5): 1511-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23380374

ABSTRACT

The onset of resistance to approved anti-AIDS drugs by HIV necessitates the search for novel inhibitors of HIV-1 reverse transcriptase (RT). Developing single molecular agents concurrently occupying the nucleoside and nonnucleoside binding sites in RT is an intriguing idea but the proof of concept has so far been elusive. As a first step, we describe molecular modeling to guide focused chemical syntheses of conjugates having nucleoside (d4T) and nonnucleoside (TIBO) moieties tethered by a flexible polyethylene glycol (PEG) linker. A triphosphate of d4T-6PEG-TIBO conjugate was successfully synthesized that is recognized as a substrate by HIV-1 RT and incorporated into a double-stranded DNA.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/enzymology , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Binding Sites , Drug Design , HIV Infections/drug therapy , HIV Reverse Transcriptase/chemistry , Humans , Models, Molecular , Nucleosides/chemistry , Nucleosides/pharmacology , Polyethylene Glycols/chemistry , Reverse Transcriptase Inhibitors/metabolism
7.
Chem Biol Drug Des ; 80(6): 971-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22978415

ABSTRACT

DOT1L is the human protein methyltransferase responsible for catalyzing the methylation of histone H3 on lysine 79 (H3K79). The ectopic activity of DOT1L, associated with the chromosomal translocation that is a universal hallmark of MLL-rearranged leukemia, is a required driver of leukemogenesis in this malignancy. Here, we present studies on the structure-activity relationship of aminonucleoside-based DOT1L inhibitors. Within this series, we find that improvements in target enzyme affinity and selectivity are driven entirely by diminution of the dissociation rate constant for the enzyme-inhibitor complex, leading to long residence times for the binary complex. The biochemical K(i) and residence times measured for these inhibitors correlate well with their effects on intracellular H3K79 methylation and MLL-rearranged leukemic cell killing. Crystallographic studies reveal a conformational adaptation mechanism associated with high-affinity inhibitor binding and prolonged residence time; these studies also suggest that conformational adaptation likewise plays a critical role in natural ligand interactions with the enzyme, hence, facilitating enzyme turnover. These results provide critical insights into the role of conformational adaptation in the enzymatic mechanism of catalysis and in pharmacologic intervention for DOT1L and other members of this enzyme class.


Subject(s)
Enzyme Inhibitors/chemistry , Methyltransferases/antagonists & inhibitors , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/metabolism , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Kinetics , Methylation , Methyltransferases/metabolism , Molecular Docking Simulation , Nucleosides/chemistry , Nucleosides/metabolism , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
8.
J Virol ; 86(13): 7052-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532675

ABSTRACT

A major goal of AIDS vaccine development is to design vaccination strategies that can elicit broad and potent protective antibodies. The initial viral targets of neutralizing antibodies (NAbs) early after human or simian immunodeficiency virus (HIV/SIV) infection are not known. The identification of early NAb epitopes that induce protective immunity or retard the progression of disease is important for AIDS vaccine development. The aim of this study was to determine the Env residues targeted by early SIV NAbs and to assess the influence of prior vaccination on neutralizing antibody kinetics and specificity during early infection. We previously described stereotypic env sequence variations in SIVmac251-infected rhesus monkeys that resulted in viral escape from NAbs. Here, we defined the early viral targets of neutralization and determined whether the ability of serum antibody from infected monkeys to neutralize SIV was altered in the setting of prior vaccination. To localize the viral determinants recognized by early NAbs, a panel of mutant pseudoviruses was assessed in a TZM-bl reporter gene neutralization assay to define the precise changes that eliminate recognition by SIV Env-specific NAbs in 16 rhesus monkeys. Changing R420 to G or R424 to Q in V4 of Env resulted in the loss of recognition by NAbs in vaccinated monkeys. In contrast, mutations in the V1 region of Env did not alter the NAb profile. These findings indicate that early NAbs are directed toward SIVmac251 Env V4 but not the V1 region, and that this env vaccination regimen did not alter the kinetics or the breadth of NAbs during early infection.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Epitopes, B-Lymphocyte/immunology , Gene Products, env/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Epitopes, B-Lymphocyte/genetics , Gene Products, env/genetics , Macaca mulatta , Male , Mutant Proteins/genetics , Mutant Proteins/immunology , Neutralization Tests , Simian Immunodeficiency Virus/genetics
9.
Anal Biochem ; 416(2): 206-10, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21669181

ABSTRACT

Jump dilution analysis is commonly used to evaluate the reversibility of inhibition and to quantify the residence time of the inhibitor-enzyme complex. During hit and lead characterization, one sometimes observes apparently linear progress curves after jump dilution that display activity recoveries that are intermediate between those expected for fully reversible and irreversible inhibition. Computer simulations of progress curves after jump dilution indicate that seemingly linear progress curves can result when dealing with tight-binding inhibitors if substoichiometric concentrations of inhibitor are preincubated with enzyme. In this situation, the activity recovered is comparable to that expected for instantaneously reversible inhibitors. In addition, simulations demonstrate that intermediate values of activity recovery may be observed for compounds with modestly slow dissociation rates (i.e., residence times >0min but ≤20min) when the attending curvature of the data is not accounted for. The observation of intermediate values of recovery can, thus, serve as an indication of either modest residence time or a contaminating inactivator within an inhibitor sample, in either case prompting greater scrutiny of the test compound.


Subject(s)
Enzymes/chemistry , Enzyme Inhibitors/chemistry , Enzymes/metabolism , Kinetics , Protein Binding , Time Factors
10.
J Virol ; 84(2): 953-63, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19906933

ABSTRACT

The evolution of envelope mutations by replicating primate immunodeficiency viruses allows these viruses to escape from the immune pressure mediated by neutralizing antibodies. Vaccine-induced anti-envelope antibody responses may accelerate and/or alter the specificity of the antibodies, thus shaping the evolution of envelope mutations in the replicating virus. To explore this possibility, we studied the neutralizing antibody response and the envelope sequences in rhesus monkeys vaccinated with either gag-pol-nef immunogens or gag-pol-nef immunogens in combination with env and then infected with simian immunodeficiency virus (SIV). Using a pseudovirion neutralization assay, we demonstrate that envelope vaccination primed for an accelerated neutralizing antibody response following virus challenge. To monitor viral envelope evolution in these two cohorts of monkeys, full-length envelopes from plasma virus isolated at weeks 37 and 62 postchallenge were sequenced by single genome amplification to identify sites of envelope mutations. We show that env vaccination was associated with a change in the pattern of envelope mutations. Prevalent mutations in sequences from gag-pol-nef vaccinees included deletions in both variable regions 1 and 4 (V1 and V4), whereas deletions in the env vaccinees occurred only in V1. These data show that env vaccination altered the focus of the antibody-mediated selection pressure on the evolution of envelope following SIV challenge.


Subject(s)
Evolution, Molecular , Macaca mulatta/virology , SAIDS Vaccines , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Viral Envelope Proteins , Amino Acid Sequence , Animals , Antibodies, Neutralizing , Cell Line , Humans , Molecular Sequence Data , Mutation , Neutralization Tests , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Vaccination , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
11.
Bioorg Med Chem ; 16(24): 10270-80, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18996020

ABSTRACT

Several novel thiourea derivatives of the NNRTI HI-236 substituted at the C-2 oxygen of the phenyl ring have been synthesized and evaluated for their inhibitory activity against HIV-1 (IIIB) replication in MT-2 cell cultures. The compounds were synthesized in order to fine-tune the activity of HI-236 as well as to gain insight into spatial characteristics in the pocket pertaining to the positional choice of tether in the design of [NRTI]-tether-[HI-236] bifunctional inhibitors. Two of the thiourea derivatives bearing a butynyl (6c) or hydroxyethyl tether (6n) were endowed with improved anti-HIV activity compared to HI-236. NNRTI activity was confirmed by a cell-free RT assay on six of the derivatives in which 6c returned an IC(50) of 3.8 nM compared to 28 nM for HI-236, establishing it as an improved lead for HI-236. The structure-activity profile is discussed in terms of potential interactions in the NNRTI pocket as suggested by a docking model using AutoDock, which have a bearing on the bifunctional drug design.


Subject(s)
Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Pyridines/chemistry , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacology , Thiourea/analogs & derivatives , Anti-HIV Agents/chemistry , Cells, Cultured , Computer Simulation , Drug Design , HIV-1/enzymology , Inhibitory Concentration 50 , Molecular Structure , Reverse Transcriptase Inhibitors/chemistry , Structure-Activity Relationship , Thiourea/chemistry
12.
FASEB J ; 21(14): 3795-808, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17639073

ABSTRACT

The HIV/AIDS pandemic has existed for >25 years. Extensive work globally has provided avenues to combat viral infection, but the disease continues to rage on in the human population and infected approximately 4 million people in 2006 alone. In this review, we provide a brief history of HIV/AIDS, followed by analysis of one therapeutic target of HIV-1: its reverse transcriptase (RT). We discuss the biochemical characterization of RT in order to place emphasis on possible avenues of inhibition, which now includes both nucleoside and non-nucleoside modalities. Therapies against RT remain a cornerstone of anti-HIV treatment, but the virus eventually resists inhibition through the selection of drug-resistant RT mutations. Current inhibitors and associated resistance are discussed, with the hopes that new therapeutics can be developed against RT.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/enzymology , HIV Infections/virology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , HIV-1/enzymology , Reverse Transcriptase Inhibitors/therapeutic use , HIV Infections/drug therapy , HIV Infections/epidemiology , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Humans
13.
Antivir Chem Chemother ; 18(2): 83-92, 2007.
Article in English | MEDLINE | ID: mdl-17542153

ABSTRACT

(-)-(2R,4R)-1-(2-Hydroxymethyl-1,3-dioxolan-4yl)thymine (DOT) is a thymidine analogue that has potent in vitro activity against wild-type and nucleoside reverse transcriptase inhibitor (NRTI)-resistant HIV. For nucleoside analogues to inhibit viral replication, they must be metabolized to the active triphosphate, which inhibits the viral reverse transcriptase (RT). Using purified enzymes, the kinetics of DOT phosphorylation, inhibition of wild-type and drug-resistant HIV-1 reverse transcriptase activity, and excision of DOT-5'-monophosphate (DOT-MP) from a chain-terminated primer were examined. DOT was phosphorylated by human thymidine kinase-1 (TK-1) but not by other pyrimidine nucleoside kinases, including the mitochondrial thymidine kinase (TK-2). Resistance to NRTIs involves decreased binding/incorporation and/or increased excision of the chain-terminating NRTI. RTs containing the D67N/K70R/T215Y/K219Q or T695-SS/T215Y mutations show enhanced removal of DOT-MP from terminated primer as well as approximately four-fold decreased binding/incorporation. The Q151M and K65R mutations appear to cause decreased inhibition by DOT-TP. However, both the K65R and Q151M mutations show decreased excision, which would confer greater stability on the terminated primer. These opposing mechanisms could offset the overall resistance profile and susceptibility. Little or no resistance was observed with the enzymes harbouring mutations resistant to lamivudine (M184V) and non-nucleoside RT inhibitors (K103N).


Subject(s)
Anti-HIV Agents/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/pharmacology , Thymine/analogs & derivatives , Adenosine Triphosphate/pharmacology , Drug Resistance, Viral , Phosphorylation , Thymine/pharmacology
14.
Bioorg Med Chem Lett ; 17(9): 2614-7, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17317163

ABSTRACT

The synthesis of bifunctional compound 10 consisting of d4U joined at C-5 to a butynyl spacer attached to HI-236 is reported using a Sonogashira coupling as a key step. As a non-cleavable bifunctional HIV inhibitor incorporating an NRTI with an NNRTI, 10 shows good inhibitory activity (EC(50)=250 nM) against HIV (IIIB) replication in MT-2 cell culture, which is eight times greater than that of d4T and between those of the two component drugs.


Subject(s)
Anti-HIV Agents/chemical synthesis , Chemistry, Pharmaceutical/methods , HIV Reverse Transcriptase/antagonists & inhibitors , Pyridines/chemistry , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacology , Thiourea/analogs & derivatives , Anti-HIV Agents/pharmacology , Cell Line , Dimerization , Drug Design , Humans , Models, Chemical , Molecular Conformation , Nucleosides/chemistry , Pyridines/pharmacology , Thiourea/chemistry , Thiourea/pharmacology , Virus Replication/drug effects
15.
Biochemistry ; 45(23): 7334-40, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16752922

ABSTRACT

Synergy between the anti-human immunodeficiency virus type 1 (HIV) nucleoside reverse transcriptase (RT) inhibitors (NRTIs) and nonnucleoside RT inhibitors (NNRTIs) results from a general mechanism in which NNRTIs inhibit ATP-mediated removal of NRTIs from chain-terminated primers by decreasing the maximum rate of removal, thus sustaining NRTI chain termination. With this molecular mechanism of synergy, beta-D-(+)-3'-azido-3'-deoxythymidine monophosphate (AZTMP) removal was examined in the context of clinically relevant RT mutants. The IC50 value for inhibition by nevirapine against wild-type (WT) RT in our removal assay was 3 microM, but this concentration had no effect on removal by the nevirapine-resistant Y181C mutant. Rather, a approximately 83-fold increase in nevirapine was required to decrease the rate of removal by 50% for this mutant. Efavirenz displayed a 100 nM IC50 value against WT and the efavirenz-sensitive Y181C mutant, but the efavirenz-resistant mutants K103N and K103N/Y181C required a 6-fold increase in efavirenz concentration to achieve the same effect. A newer generation NNRTI, TMC125, showed potency (55 nM) against WT and all mutants, paralleling the activity of this inhibitor relative to nevirapine and efavirenz in cell culture. When tested against the AZT-resistant mutant, all NNRTIs inhibited removal by greater than 50%, showing that this mutant is hypersensitive to NNRTIs. Altogether these results illustrate that both the NNRTI and NRTI mutations can modulate chain termination. This demonstrates that sustaining synergistic HIV inhibition in combination NRTI/NNRTI therapy requires NNRTIs that are potent against WT virus and possess favorable activity profiles against clinically relevant mutations.


Subject(s)
HIV Reverse Transcriptase/genetics , HIV-1/enzymology , Mutation , Reverse Transcriptase Inhibitors/pharmacology , Base Sequence , DNA Primers
16.
Curr Pharm Des ; 12(15): 1857-65, 2006.
Article in English | MEDLINE | ID: mdl-16724952

ABSTRACT

To date three nonnucleoside reverse transcriptase inhibitors (NNRTIs) have been approved by the U.S. Food and Drug Administration for the treatment of human immunodeficiency virus type 1 infection. A limiting factor in the effectiveness of these agents is the development of resistance, manifested by amino acid substitutions within the virally encoded reverse transcriptase (RT). Understanding the mechanism of action of these agents and how resistance develops have broadened the field of NNRTI research to elucidate structural and biochemical features of inhibition in hopes of creating better inhibitors. In this review, the history of NNRTIs will preface the many studies characterizing inhibition and the development of a new paradigm for understanding the molecular mechanism of drug resistance to NNRTIs. Combination therapies including nonnucleoside inhibitors will be discussed, concluding with remarks on potential new inhibitors.


Subject(s)
Drug Design , HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/chemistry , Anti-HIV Agents/chemistry , Anti-HIV Agents/therapeutic use , Drug Resistance, Viral/genetics , HIV Infections/drug therapy , HIV Infections/virology , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , Humans , Models, Molecular , Molecular Structure , Reverse Transcriptase Inhibitors/therapeutic use
17.
Bioorg Med Chem Lett ; 16(3): 668-71, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16298131

ABSTRACT

Following computational analyses, potential non-nucleoside inhibitors of HIV-1 reverse transcriptase have been pursued through synthesis and assaying for anti-viral activity. The general class Het-NH-Ph-U has been considered, where Het is an aromatic heterocycle and U is an unsaturated, hydrophobic group. Results for compounds with Het=2-thiazoyl and 2-pyrimidinyl are the focus of this report.


Subject(s)
Amines/chemistry , HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/chemical synthesis , Amines/pharmacology , Drug Resistance, Multiple, Viral , HIV Infections/drug therapy , HIV Infections/virology , Heterocyclic Compounds/chemistry , Hydrophobic and Hydrophilic Interactions , Pyrimidines/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Structure-Activity Relationship , Thiazoles/chemistry
18.
Bioorg Med Chem Lett ; 16(3): 663-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16263277

ABSTRACT

Design principles are delineated for non-nucleoside inhibitors for HIV-1 reverse transcriptase (NNRTIs). Simultaneous optimization of binding affinity for wild-type RT, tolerance for viral mutations, and physical properties is pursued. Automated lead generation with the growing program BOMB, Monte Carlo simulations with free-energy perturbation theory for lead optimization, and property analysis with QikProp are featured. An initial 30 microM lead has been optimized rapidly to the 10 nM level.


Subject(s)
Computer-Aided Design , HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/pharmacology , Drug Resistance, Multiple, Viral , HIV Infections/drug therapy , HIV Infections/virology , HIV Reverse Transcriptase/genetics , Models, Molecular , Molecular Structure , Monte Carlo Method , Mutation , Reverse Transcriptase Inhibitors/chemistry
19.
Antiviral Res ; 67(1): 10-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15950748

ABSTRACT

A novel non-chain terminating nucleoside analog anti-HIV inhibitor, KP-1212 has been designed to form base pairs with multiple bases that may lead to mutagenesis in the HIV-1 viral genome. After multiple replication cycles, the accumulation of mutations surpasses a crucial threshold beyond which the virus can no longer replicate. HIV-1 reverse transcriptase (RT) incorporates the KP-1212 monophosphate into the genome during viral replication after metabolic activation of the KP-1212 nucleoside to the triphosphate. The propensity for forming alternate base pairs with the KP-1212 nucleotide leads to mismatched nucleotides and the subsequent misincorporation is the basis for the inhibitory activity. The results showed that HIV-1 RT and human mitochondrial DNA polymerase (Pol gamma) incorporated KP-1212-TP with a significant level of efficiency, whereas mouse DNA polymerase beta (Pol beta) did not. Misincorporation studies suggest that both HIV-1 RT and Pol gamma may cause mutations at significantly high rates. These in vitro data confirm the mechanistic basis of KP-1212 as a viral mutagen but suggest that there may be a potential for toxicity to the mitochondria.


Subject(s)
Anti-HIV Agents/metabolism , DNA-Directed DNA Polymerase/metabolism , Deoxycytosine Nucleotides/metabolism , HIV Reverse Transcriptase/metabolism , Mutation , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Binding, Competitive , DNA-Directed DNA Polymerase/drug effects , Deoxycytosine Nucleotides/chemistry , Deoxycytosine Nucleotides/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/drug effects , Humans , Mitochondria/enzymology , Nucleic Acid Synthesis Inhibitors
20.
J Biol Chem ; 279(8): 6221-4, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14722107

ABSTRACT

Combination therapies treating human immunodeficiency virus type 1 (HIV-1) infection delay the emergence of drug-resistant virus and exhibit synergistic inhibition. This synergy is observed within the two classes of inhibitors that target the essential viral reverse transcriptase (RT): the chain-terminating nucleoside analogs (NRTIs) and the allosteric nonnucleosides (NNRTIs) that bind in a pocket distinct from the active site. A general mechanism to define the molecular basis for synergy between these two classes remains to be elucidated. Previous mechanistic studies from our laboratory (Spence, R. A., Kati, W. M., Anderson, K. S., and Johnson, K. A. (1995) Science 267, 988-993) have shown that the natural deoxynucleoside triphosphate and the NNRTI can simultaneously bind to their respective sites. This work also suggests communication between the two sites, since the inhibition of RT by NNRTIs is manifested through a remote effect on the chemical step. This interplay between the two sites offers a plausible hypothesis for understanding synergy in which binding of NNRTIs modulates the chain termination by NRTIs. The present study supports this hypothesis by illustrating that the clinically approved NNRTIs, nevirapine and efavirenz, inhibit the ATP-mediated removal of AZTMP, d4TMP, ddCMP, (-)3TCMP, (-)FTCMP, and (+)3TCMP, thereby prolonging the effectiveness of chain termination. This inhibition is mediated through an effect on both the rate of the chemical step and binding of ATP, resulting in an overall decrease in efficiency of removal. This work substantiates communication between the two binding pockets, the sustained use of combination therapy to treat HIV infection, and a molecular basis for understanding synergy.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/chemistry , HIV Infections/drug therapy , Nucleosides/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/pharmacology , Allosteric Site , Binding Sites , Dose-Response Relationship, Drug , Drug Resistance, Viral , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Kinetics , Models, Molecular , Nevirapine/pharmacology , Oligonucleotides/chemistry , Protein Binding , Reverse Transcriptase Inhibitors/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...