Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nature ; 584(7822): 614-618, 2020 08.
Article in English | MEDLINE | ID: mdl-32612233

ABSTRACT

Oral antiretroviral agents provide life-saving treatments for millions of people living with HIV, and can prevent new infections via pre-exposure prophylaxis1-5. However, some people living with HIV who are heavily treatment-experienced have limited or no treatment options, owing to multidrug resistance6. In addition, suboptimal adherence to oral daily regimens can negatively affect the outcome of treatment-which contributes to virologic failure, resistance generation and viral transmission-as well as of pre-exposure prophylaxis, leading to new infections1,2,4,7-9. Long-acting agents from new antiretroviral classes can provide much-needed treatment options for people living with HIV who are heavily treatment-experienced, and additionally can improve adherence10. Here we describe GS-6207, a small molecule that disrupts the functions of HIV capsid protein and is amenable to long-acting therapy owing to its high potency, low in vivo systemic clearance and slow release kinetics from the subcutaneous injection site. Drawing on X-ray crystallographic information, we designed GS-6207 to bind tightly at a conserved interface between capsid protein monomers, where it interferes with capsid-protein-mediated interactions between proteins that are essential for multiple phases of the viral replication cycle. GS-6207 exhibits antiviral activity at picomolar concentrations against all subtypes of HIV-1 that we tested, and shows high synergy and no cross-resistance with approved antiretroviral drugs. In phase-1 clinical studies, monotherapy with a single subcutaneous dose of GS-6207 (450 mg) resulted in a mean log10-transformed reduction of plasma viral load of 2.2 after 9 days, and showed sustained plasma exposure at antivirally active concentrations for more than 6 months. These results provide clinical validation for therapies that target the functions of HIV capsid protein, and demonstrate the potential of GS-6207 as a long-acting agent to treat or prevent infection with HIV.


Subject(s)
Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Capsid Proteins/antagonists & inhibitors , HIV-1/drug effects , Adolescent , Adult , Anti-HIV Agents/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cells, Cultured , Drug Resistance, Viral/genetics , Female , HIV-1/growth & development , Humans , Male , Middle Aged , Models, Molecular , Virus Replication/drug effects , Young Adult
2.
Bioorg Med Chem Lett ; 28(3): 541-546, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29254643

ABSTRACT

We hereby disclose the discovery of inhibitors of CaMKII (7h and 7i) that are highly potent in rat ventricular myocytes, selective against hERG and other off-target kinases, while possessing good CaMKII tissue isoform selectivity (cardiac γ/δ vs. neuronal α/ß). In vitro and in vivo ADME/PK studies demonstrated the suitability of these CaMKII inhibitors for PO (7h rat F = 73%) and IV pharmacological studies.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
3.
J Mol Cell Cardiol ; 94: 122-130, 2016 05.
Article in English | MEDLINE | ID: mdl-27066997

ABSTRACT

AIMS: Increases in late Na(+) current (late INa) and activation of Ca(2+)/calmodulin-dependent protein kinase (CaMKII) are associated with atrial arrhythmias. CaMKII also phosphorylates Nav1.5, further increasing late INa. The combination of a CaMKII inhibitor with a late INa inhibitor may be superior to each compound alone to suppress atrial arrhythmias. Therefore, we investigated the effect of a CaMKII inhibitor in combination with a late INa inhibitor on anemone toxin II (ATX-II, a late INa enhancer)-induced atrial arrhythmias. METHODS AND RESULTS: Rat right atrial tissue was isolated and preincubated with either the CaMKII inhibitor autocamtide-2-related inhibitory peptide (AIP), the late INa inhibitor GS458967, or both, and then exposed to ATX-II. ATX-II increased diastolic tension and caused fibrillation of isolated right atrial tissue. AIP (0.3µmol/L) and 0.1µmol/L GS458967 alone inhibited ATX-II-induced arrhythmias by 20±3% (mean±SEM, n=14) and 34±5% (n=13), respectively, whereas the two compounds in combination inhibited arrhythmias by 81±4% (n=10, p<0.05, vs either AIP or GS458967 alone or the calculated sum of individual effects of both compounds). AIP and GS458967 also attenuated the ATX-induced increase of diastolic tension. Consistent with the mechanical and electrical data, 0.3µmol/L AIP and 0.1µmol/L GS458967 each inhibited ATX-II-induced CaMKII phosphorylation by 23±3% and 32±4%, whereas the combination of both compounds inhibited CaMKII phosphorylation completely. CONCLUSION: The effects of an enhanced late INa to induce arrhythmic activity and activation of CaMKII in atria are attenuated synergistically by inhibitors of late INa and CaMKII.


Subject(s)
Action Potentials , Ataxin-2/metabolism , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Sodium/metabolism , Animals , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Heart Atria/drug effects , Heart Atria/metabolism , Male , Myocardial Contraction/drug effects , Rats , Sulfonamides/pharmacology
4.
Antimicrob Agents Chemother ; 60(3): 1847-1853, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26824950

ABSTRACT

Ledipasvir (LDV; GS-5885), a component of Harvoni (a fixed-dose combination of LDV with sofosbuvir [SOF]), is approved to treat chronic hepatitis C virus (HCV) infection. Here, we report key preclinical antiviral properties of LDV, including in vitro potency, in vitro resistance profile, and activity in combination with other anti-HCV agents. LDV has picomolar antiviral activity against genotype 1a and genotype 1b replicons with 50% effective concentration (EC50) values of 0.031 nM and 0.004 nM, respectively. LDV is also active against HCV genotypes 4a, 4d, 5a, and 6a with EC50 values of 0.11 to 1.1 nM. LDV has relatively less in vitro antiviral activity against genotypes 2a, 2b, 3a, and 6e, with EC50 values of 16 to 530 nM. In vitro resistance selection with LDV identified the single Y93H and Q30E resistance-associated variants (RAVs) in the NS5A gene; these RAVs were also observed in patients after a 3-day monotherapy treatment. In vitro antiviral combination studies indicate that LDV has additive to moderately synergistic antiviral activity when combined with other classes of HCV direct-acting antiviral (DAA) agents, including NS3/4A protease inhibitors and the nucleotide NS5B polymerase inhibitor SOF. Furthermore, LDV is active against known NS3 protease and NS5B polymerase inhibitor RAVs with EC50 values equivalent to those for the wild type.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Fluorenes/pharmacology , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Viral Nonstructural Proteins/antagonists & inhibitors , Carrier Proteins/antagonists & inhibitors , Cell Line, Tumor , Drug Combinations , Drug Resistance, Viral/genetics , Drug Synergism , Genotype , HeLa Cells , Hepacivirus/genetics , Humans , Intracellular Signaling Peptides and Proteins , Microbial Sensitivity Tests , Sofosbuvir/pharmacology
5.
Biochemistry ; 54(13): 2240-8, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25774576

ABSTRACT

HIV capsid protein is an important target for antiviral drug design. High-throughput screening campaigns have identified two classes of compounds (PF74 and BI64) that directly target HIV capsid, resulting in antiviral activity against HIV-1 and HIV-2 laboratory strains. Using recombinant proteins, we developed a suite of label-free assays to mechanistically understand how these compounds modulate capsid activity. PF74 preferentially binds to the preassembled hexameric capsid form and prevents disruption of higher-order capsid structures by stabilizing capsid intersubunit interactions. BI64 binds only the monomeric capsid and locks the protein in the assembly incompetent monomeric form by disrupting capsid intersubunit interactions. We also used these assays to characterize the interaction between capsid and the host protein cleavage and polyadenylation specific factor 6 (CPSF6). Consistent with recently published results, our assays revealed CPSF6 activates capsid polymerization and preferentially binds to the preassembled hexameric capsid form similar to the small molecule compound, PF74. Furthermore, these label-free assays provide a robust method for facilitating the identification of a different class of small molecule modulators of capsid function.


Subject(s)
Anti-HIV Agents/pharmacology , Biosensing Techniques/methods , Capsid/metabolism , Drug Evaluation, Preclinical/methods , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Sequence , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Benzimidazoles/pharmacology , Capsid/chemistry , HIV-1 , Host-Pathogen Interactions/drug effects , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacology , Molecular Sequence Data , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/metabolism , Phenylalanine/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Small Molecule Libraries/pharmacology , mRNA Cleavage and Polyadenylation Factors/genetics
6.
Antimicrob Agents Chemother ; 58(1): 386-96, 2014.
Article in English | MEDLINE | ID: mdl-24165192

ABSTRACT

One of the most challenging goals of hepatitis C virus (HCV) research is to develop well-tolerated regimens with high cure rates across a variety of patient populations. Such a regimen will likely require a combination of at least two distinct direct-acting antivirals (DAAs). Combining two or more DAAs with different resistance profiles increases the number of mutations required for viral breakthrough. Currently, most DAAs inhibit HCV replication. We recently reported that the combination of two distinct classes of HCV inhibitors, entry inhibitors and replication inhibitors, prolonged reductions in extracellular HCV in persistently infected cells. We therefore sought to identify new inhibitors targeting aspects of the HCV replication cycle other than RNA replication. We report here the discovery of the first small-molecule HCV infectivity inhibitor, GS-563253, also called HCV infectivity inhibitor 1 (HCV II-1). HCV II-1 is a substituted tetrahydroquinoline that selectively inhibits genotype 1 and 2 HCVs with low-nanomolar 50% effective concentrations. It was identified through a high-throughput screen and subsequent chemical optimization. HCV II-1 only permits the production and release of noninfectious HCV particles from cells. Moreover, infectious HCV is rapidly inactivated in its presence. HCV II-1 resistance mutations map to HCV E2. In addition, HCV-II prevents HCV endosomal fusion, suggesting that it either locks the viral envelope in its prefusion state or promotes a viral envelope conformation change incapable of fusion. Importantly, the discovery of HCV II-1 opens up a new class of HCV inhibitors that prolong viral suppression by HCV replication inhibitors in persistently infected cell cultures.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Antiviral Agents/chemistry , Cell Line , Drug Resistance, Viral , Hepacivirus/metabolism , Hepatitis C/drug therapy , Humans , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
7.
J Nat Prod ; 77(3): 618-24, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24495105

ABSTRACT

An extract of Humicola fuscoatra (UCSC strain no. 108111A) was shown to reactivate latent HIV-1 expression in an in vitro model of central memory CD4+ T cells. We report the bioassay-guided isolation and structure determination of several resorcyclic acid lactones, including four known compounds, radicicol (1, aka. monorden) and pochonins B (2), C (3), and N (4), and three new analogues, radicicols B-D (5-7). Compounds 1-3 and 5 showed moderate activities in the memory T cell model of HIV-1 latency. Radicicol (1) displayed lower potency in reactivating latent HIV-1 (EC50 = 9.1 µM) relative to the HDAC inhibitors apicidin (EC50 = 0.3 µM), romidepsin (EC50 = 0.003 µM), and SAHA (EC50 = 0.6 µM); however, it achieved equivalent maximum efficacy relative to the positive control compounds (98% of SAHA and romidepsin).


Subject(s)
Ascomycota/chemistry , Biological Products/pharmacology , CD4-Positive T-Lymphocytes/virology , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Lactones/chemistry , Macrolides/pharmacology , Biological Products/chemistry , HIV Infections/virology , Histone Deacetylase Inhibitors/chemistry , Humans , Lactones/pharmacology , Macrolides/chemistry , Marine Biology , Models, Biological , Molecular Structure , Virus Latency/drug effects
8.
Arch Biochem Biophys ; 506(2): 130-6, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21081101

ABSTRACT

Many of the cellular responses to Ca++ signaling are modulated by a family of multifunctional Ca++/calmodulin dependent protein kinases (CaMKs): CaMK I, CaMK II and CaMK IV. In order to further understand the role of CaMKs, we investigated the kinetic mechanism of CaMK II isozymes in comparison with those of CaMK I and CaMK IV by analyzing their steady state kinetics using phospholamban as a phosphoacceptor. The results indicated that (a) the CaMK family's reaction mechanisms were of the sequential type in which all substrates must bind to enzyme before any product is released; (b) CaMK I and CaMK IV exhibited random sequential mechanism where either phospholamban or ATP can bind to the free enzyme; (c) the data of product inhibition for CaMK IIs best fit with an Ordered Bi Bi mechanism in which phospholamban is the first substrate to bind and ADP is the last product to be released; and (d) the constant α (ratio of apparent dissociation constants for binding peptide in the presence and absence of the second ligand) of all isozymes for ATP and peptide was higher than 1 indicating that the binding of phospholamban to CaMK decreased the enzyme's affinity toward ATP.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 1/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 1/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 4/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 4/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Humans , In Vitro Techniques , Kinetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
9.
Bioorg Med Chem Lett ; 21(5): 1447-51, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21300545

ABSTRACT

The synthesis and preliminary studies of the SAR of novel 3,5-diarylazole inhibitors of Protein Kinase D (PKD) are reported. Notably, optimized compounds in this class have been found to be active in cellular assays of phosphorylation-dependant HDAC5 nuclear export, orally bioavailable, and highly selective versus a panel of additional putative histone deacetylase (HDAC) kinases. Therefore these compounds could provide attractive tools for the further study of PKD/HDAC5 signaling.


Subject(s)
Azoles/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Azoles/chemical synthesis , Azoles/chemistry , Azoles/pharmacokinetics , Biological Availability , Histone Deacetylases/metabolism , Inhibitory Concentration 50 , Molecular Structure , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Rats , Rats, Sprague-Dawley , Signal Transduction , Structure-Activity Relationship
10.
J Biol Chem ; 284(48): 33580-99, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19801648

ABSTRACT

The interaction between lens epithelium-derived growth factor/transcriptional co-activator p75 (LEDGF) and human immunodeficiency virus type 1 (HIV-1) integrase (IN) is essential for HIV-1 replication. Homogeneous time-resolved fluorescence resonance energy transfer assays were developed to characterize HIV-1 integrase dimerization and the interaction between LEDGF and IN dimers. Using these assays in an equilibrium end point dose-response format with mathematical modeling, we determined the dissociation constants of IN dimers (K(dimer) = 67.8 pm) and of LEDGF from IN dimers (K(d) = 10.9 nm). When used in a kinetic format, the assays allowed the determination of the on- and off-rate constants for these same interactions. Integrase dimerization had a k(on) of 0.1247 nm(-1) x min(-1) and a k(off) of 0.0080 min(-1) resulting in a K(dimer) of 64.5 pm. LEDGF binding to IN dimers had a k(on) of 0.0285 nm(-1).min(-1) and a k(off) of 0.2340 min(-1) resulting in a K(d) of 8.2 nm. These binding assays can also be used in an equilibrium end point competition format. In this format, the IN catalytic core domain produced a K(i) of 15.2 nm while competing for integrase dimerization, confirming the very tight interaction of IN with itself. In the same format, LEDGF produced a K(i) value of 35 nm when competing for LEDGF binding to IN dimers. In summary, this study describes a methodology combining homogeneous time-resolved fluorescence resonance energy transfer and mathematical modeling to derive the affinities between IN monomers and between LEDGF and IN dimers. This study revealed the significantly tighter nature of the IN-IN dimer compared with the IN-LEDGF interaction.


Subject(s)
HIV Integrase/chemistry , HIV Integrase/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Algorithms , Amino Acid Sequence , Binding, Competitive , Fluorescence Resonance Energy Transfer , HIV Integrase/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Kinetics , Models, Biological , Models, Chemical , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Multimerization
11.
Biochim Biophys Acta ; 1789(5): 422-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19414071

ABSTRACT

Histone deacetylase 5 (HDAC5) represses expression of nuclear genes that promote cardiac hypertrophy. Agonism of a variety of G protein coupled receptors (GPCRs) triggers phosphorylation-dependent nuclear export of HDAC5 via the CRM1 nuclear export receptor, resulting in derepression of pro-hypertrophic genes. A cell-based high-throughput screen of a commercial compound collection was employed to identify compounds with the ability to preserve the nuclear fraction of GFP-HDAC5 in primary cardiomyocytes exposed to GPCR agonists. A hit compound potently inhibited agonist-induced GFP-HDAC5 nuclear export in cultured neonatal rat ventricular myocytes (NRVMs). A small set of related compounds was designed and synthesized to evaluate structure-activity relationship (SAR). The results demonstrated that inhibition of HDAC5 nuclear export was a result of compounds irreversibly reacting with a key cysteine residue in CRM1 that is required for its function. CRM1 inhibition by the compounds also resulted in potent suppression of cardiomyocyte hypertrophy. These studies define a novel class of anti-hypertrophic compounds that function through irreversible inhibition of CRM1-dependent nuclear export.


Subject(s)
Cardiomegaly/drug therapy , Histone Deacetylases/metabolism , Karyopherins/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Amides/pharmacology , Aniline Compounds/pharmacology , Animals , Cardiomegaly/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Histone Deacetylase Inhibitors , Histone Deacetylases/chemistry , Humans , Karyopherins/metabolism , Microscopy, Fluorescence , Myocytes, Cardiac/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Exportin 1 Protein
12.
Antimicrob Agents Chemother ; 54(8): 3099-106, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20516274

ABSTRACT

The hepatitis C virus (HCV) subgenomic replicon is the primary tool for evaluating the activity of anti-HCV compounds in drug discovery research. Despite the prevalence of HCV genotype 1a (approximately 70% of U.S. HCV patients), few genotype 1a reporter replicon cell lines have been described; this is presumably due to the low replication capacity of such constructs in available Huh-7 cells. In this report, we describe the selection of highly permissive Huh-7 cell lines that support robust replication of genotype 1a subgenomic replicons harboring luciferase reporter genes. These novel cell lines support the replication of multiple genotype 1a replicons (including the H77 and SF9 strains), are significantly more permissive to genotype 1a HCV replication than parental Huh7-Lunet cells, and maintain stable genotype 1a replication levels suitable for antiviral screening. We found that the sensitivity of genotype 1a luciferase replicons to known antivirals was highly consistent between individual genotype 1a clonal cell lines but could vary significantly between genotypes 1a and 1b. Sequencing of the nonstructural region of 12 stable replicon cell clones suggested that the enhanced permissivity is likely due to cellular component(s) in these new cell lines rather than the evolution of novel adaptive mutations in the replicons. These new reagents will enhance drug discovery efforts targeting genotype 1a and facilitate the profiling of compound activity among different HCV genotypes and subtypes.


Subject(s)
Antiviral Agents/pharmacology , Genes, Reporter , Hepacivirus/classification , Hepacivirus/drug effects , Replicon/genetics , Cell Line, Tumor , Genotype , Hepacivirus/genetics , Hepacivirus/physiology , Humans , Luciferases/genetics , Luciferases/metabolism , Microbial Sensitivity Tests , RNA, Viral/genetics , RNA, Viral/metabolism , Replicon/physiology , Virus Replication/drug effects
13.
SLAS Discov ; 23(9): 919-929, 2018 10.
Article in English | MEDLINE | ID: mdl-30011241

ABSTRACT

Bruton's tyrosine kinase (BTK) is a clinically validated target for B-cell leukemias and lymphomas with FDA-approved small-molecule inhibitors ibrutinib and acalabrutinib. Tirabrutinib (GS-4059/ONO-4059, Gilead Sciences, Inc., Foster City, CA) is a second-generation, potent, selective, irreversible BTK inhibitor in clinical development for lymphoid malignancies, including chronic lymphocytic leukemia (CLL) and diffuse large B-cell lymphoma (DLBCL). An accurate pharmacodynamic assay to assess tirabrutinib target coverage in phase 1/2 clinical studies will inform dose and schedule selection for advanced clinical evaluation. We developed a novel duplex homogeneous BTK occupancy assay based on time-resolved fluorescence resonance energy transfer (TR-FRET) to measure free and total BTK levels in a multiplexed format. The dual-wavelength emission property of terbium-conjugated anti-BTK antibody served as the energy donor for two fluorescent energy acceptors with distinct excitation and emission spectra. The assay was characterized and qualified using full-length purified recombinant human BTK protein and peripheral blood mononuclear cells derived from healthy volunteers and patients with CLL. We demonstrated assay utility using cells derived from lymph node and bone marrow samples from patients with CLL and DLBCL. Our TR-FRET-based BTK occupancy assay provides accurate, quantitative assessment of BTK occupancy in the clinical trial program for tirabrutinib and is in use in ongoing clinical studies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Biological Assay , Imidazoles/pharmacology , Pyrimidines/pharmacology , Biological Assay/methods , Biological Assay/standards , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Line, Tumor , Drug Stability , Enzyme Activation/drug effects , Humans , Imidazoles/chemistry , Leukemia, Lymphocytic, Chronic, B-Cell , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Reproducibility of Results
14.
J Biomol Screen ; 20(4): 498-507, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25425568

ABSTRACT

Kinetic analysis of antibodies is crucial in both clone selection and characterization. Historically, antibodies in supernatants from hybridomas are selected based on a solid-phase enzyme-linked immunosorbent assay (ELISA) in which the antigen is immobilized on the assay plate. ELISA selects clones based on a combination of antibody concentration in the supernatant and affinity. The antibody concentration in the supernatant can vary significantly and is typically unknown. Using the ELISA method, clones that express high levels of a low-affinity antibody can give an equivalent signal as clones that express low levels of a high-affinity antibody. As a consequence, using the ELISA method, superior clones can be overshadowed by inferior clones. In this study, we have applied Bio-Layer Interferometry to screen hybridoma clones based on disassociation rates using the OctetRED 384 platform. Using the OctetRED platform, we were able to screen 2000 clones within 24 hours and select clones containing high-affinity antibodies for further expansion and subsequent characterization. Using this method, we were able to identify several clones producing high-affinity antibodies that were missed by ELISA.


Subject(s)
Antibody Affinity , Hybridomas/immunology , Enzyme-Linked Immunosorbent Assay , Kinetics
15.
Org Lett ; 17(2): 262-5, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25560385

ABSTRACT

Screening of a marine natural products library afforded three new analogues of the tetronic acid containing polyketide abyssomicin family and identified abyssomicin 2 as a selective reactivator of latent HIV virus. Examination of the mode of action of this new latent HIV reactivating agent demonstrated that it functions via a distinct mechanism compared to that of existing reactivating agents and is effective at reactivating latent virus in a subset of primary patient cell lines.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Furans/chemistry , HIV Infections/drug therapy , HIV-1/drug effects , Polyketides/chemistry , Protein Kinase C/chemistry , Virus Latency/drug effects , Cell Line , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Molecular Structure , Protein Kinase C/metabolism , Protein Kinase C/pharmacology
16.
J Biomol Screen ; 17(8): 1050-61, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22811478

ABSTRACT

Apolipoprotein A-I (ApoA-I), a primary protein component of high-density lipoprotein (HDL), plays an important role in cholesterol metabolism mediating the formation of HDL and the efflux of cellular cholesterol from macrophage foam cells in arterial walls. Lipidation of ApoA-I is mediated by adenosine triphosphate (ATP) binding cassette A1 (ABCA1). Insufficient ABCA1 activity may lead to increased risk of atherosclerosis due to reduced HDL formation and cholesterol efflux. The standard radioactive assay for measuring cholesterol transport to ApoA-I has low throughput and poor dynamic range, and it fails to measure phospholipid transfer. We describe the development of two sensitive, nonradioactive high-throughput assays that report on the lipidation of ApoA-I: a homogeneous assay based on time-resolved fluorescence resonance energy transfer (TR-FRET) and a discontinuous assay that uses the label-free Epic platform. The TR-FRET assay employs HiLyte Fluor 647-labeled ApoA-I with N-terminal biotin bound to streptavidin-terbium. When fluorescent ApoA-I was incorporated into HDL, TR-FRET decreased proportionally to the increase in the ratio of lipids to ApoA-I, demonstrating that the assay was sensitive to the amount of lipid bound to ApoA-I. In the Epic assay, biotinylated ApoA-I was captured on a streptavidin-coated biosensor. Measured resonant wavelength shift was proportional to the amount of lipids associated with ApoA-I, indicating that the assay senses ApoA-I lipidation.


Subject(s)
Apolipoprotein A-I/metabolism , Fluorescence Resonance Energy Transfer/methods , High-Throughput Screening Assays/methods , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/agonists , ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Biotin , Cells, Cultured , Cholesterol/metabolism , Fluorescent Dyes/chemistry , Humans , Lipid Metabolism , Lipids , Lipoproteins, HDL/metabolism , Macrophages/metabolism , Streptavidin
17.
Curr Protoc Microbiol ; Chapter 17: Unit17.7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21805689

ABSTRACT

Hepatitis C virus (HCV) replicons are the primary tools for identifying and evaluating anti-HCV compounds during drug discovery research. Genotype 1a and 1b are the most common genotypes in North America and Europe. These genotypes display significant genetic divergence (∼20% at the nucleotide level and ∼14% at the amino acid level), which can translate into marked differences in drug susceptibility. Thus, it is critical that potential therapeutics be assessed against both genotypes to ensure antiviral efficacy in the broad genotype 1 patient population. This unit describes assays for screening HCV inhibitors using replicons with a focus on genotype 1a. Specific protocols are provided for screening 1a replicons that encode the Renilla luciferase gene and for replicons that do not encode exogenous reporter genes, both in 96-well (manual) and in 384-well (high-throughput) formats.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Hepacivirus/drug effects , Hepacivirus/genetics , Microbial Sensitivity Tests/methods , Replicon/drug effects , Cell Culture Techniques , Cell Line , Genes, Reporter , Genotype , Hepacivirus/metabolism , Humans , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
18.
J Biomol Screen ; 16(2): 211-20, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21297107

ABSTRACT

Hepatitis C virus (HCV) is a considerable global health problem for which new classes of therapeutics are needed. The authors developed a high-throughput assay to identify compounds that selectively block translation initiation from the HCV internal ribosome entry site (HCV IRES). Rabbit reticulocyte lysate conditions were optimized to faithfully report on authentic HCV IRES-dependent translation relative to a 5' capped mRNA control. The authors screened a library of ~430,000 small molecules for IRES inhibition, leading to ~1700 initial hits. After secondary counterscreening, the vast majority of hits proved to be luciferase and general translation inhibitors. Despite well-optimized in vitro translation conditions, in the end, the authors found no selective HCV IRES inhibitors but did discover a new scaffold of general translation inhibitor. The analysis of these molecules, as well we the finding that a large fraction of false positives resulted from off-target effects, highlights the challenges inherent in screens for RNA-specific inhibitors.


Subject(s)
Hepacivirus/genetics , Hepacivirus/metabolism , High-Throughput Screening Assays , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors , Animals , Drug Evaluation, Preclinical , Genes, Reporter , Humans , Protein Biosynthesis/genetics , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , Puromycin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Rabbits , Reproducibility of Results , Research Design , Small Molecule Libraries
19.
J Biomol Screen ; 16(7): 724-33, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21521800

ABSTRACT

Transforming growth factor ß (TGF-ß) type I receptor (activin receptor-like kinase 5, ALK5) has been identified as a promising target for fibrotic diseases. To find a novel inhibitor of ALK5, the authors performed a high-throughput screen of a library of 420,000 compounds using dephosphorylated ALK5. From primary hits of 1521 compounds, 555 compounds were confirmed. In total, 124 compounds were then selected for follow-up based on their unique structures and other properties. Repeated concentration-response testing and final interference assays of the above compounds resulted in the discovery of a structurally novel ALK5 inhibitor (compound 8) (N-(thiophen 2-ylmethyl)-3-(3,4,5 trimethoxyphenyl)imidazo[1,2ß]pyridazin 6-amine) with a low IC(50) value of 0.7 µM. Compound 8 also inhibited the TGF-ß-induced nuclear translocation of SMAD with an EC(50) value of 0.8 µM. Kinetic analysis revealed that compound 8 inhibited ALK5 via mixed-type inhibition, suggesting that it may bind to ALK5 differently than other published adenosine triphosphate site inhibitors.


Subject(s)
High-Throughput Screening Assays , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Adenosine Diphosphate/metabolism , Cell Line, Tumor , Computer Simulation , Fluorescence Resonance Energy Transfer , Fluoroimmunoassay , Humans , Kinetics , Molecular Conformation , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism , Small Molecule Libraries/pharmacology , Transforming Growth Factor beta/pharmacology
20.
J Med Chem ; 53(15): 5400-21, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20684591

ABSTRACT

A novel 2,6-naphthyridine was identified by high throughput screen (HTS) as a dual protein kinase C/D (PKC/PKD) inhibitor. PKD inhibition in the heart was proposed as a potential antihypertrophic mechanism with application as a heart failure therapy. As PKC was previously identified as the immediate upstream activator of PKD, PKD vs PKC selectivity was essential to understand the effect of PKD inhibition in models of cardiac hypertrophy and heart failure. The present study describes the modification of the HTS hit to a series of prototype pan-PKD inhibitors with routine 1000-fold PKD vs PKC selectivity. Example compounds inhibited PKD activity in vitro, in cells, and in vivo following oral administration. Their effects on heart morphology and function are discussed herein.


Subject(s)
Aminopyridines/chemical synthesis , Naphthyridines/chemical synthesis , Protein Kinase C/antagonists & inhibitors , Active Transport, Cell Nucleus , Administration, Oral , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Antihypertensive Agents/chemical synthesis , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cell Nucleus/metabolism , Histone Deacetylases/metabolism , Isoenzymes/antagonists & inhibitors , Male , Models, Molecular , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle Cells/pathology , Myocardium/metabolism , Myocardium/pathology , Naphthyridines/pharmacokinetics , Naphthyridines/pharmacology , Phosphorylation , Protein Binding , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL