Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 23(1): 35-46, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37735104

ABSTRACT

Small molecule inhibitors of Bruton's tyrosine kinase (BTK) have been approved for the treatment of multiple B-cell malignancies and are being evaluated for autoimmune and inflammatory diseases. Various BTK inhibitors (BTKi) have distinct potencies, selectivity profiles, and binding modes within the ATP-binding site. On the basis of the latter feature, BTKis can be classified into those that occupy the back-pocket, H3 pocket, and the hinge region only. Hypothesizing that differing binding modes may have differential impact on the B-cell receptor (BCR) signaling pathway, we evaluated the activities of multiple BTKis in B-cell lymphoma models in vitro and in vivo. We demonstrated that, although all three types of BTKis potently inhibited BTK-Y223 autophosphorylation and phospholipase C gamma 2 (PLCγ2)-Y1217 transphosphorylation, hinge-only binders were defective in inhibiting BTK-mediated calcium mobilization upon BCR activation. In addition, PLCγ2 activation was effectively blocked by back-pocket and H3 pocket binders but not by hinge-only binders. Further investigation using TMD8 cells deficient in Rac family small GTPase 2 (RAC2) revealed that RAC2 functioned as a bypass mechanism, allowing for residual BCR signaling and PLCγ2 activation when BTK kinase activity was fully inhibited by the hinge-only binders. These data reveal a kinase activity-independent function of BTK, involving RAC2 in transducing BCR signaling events, and provide mechanistic rationale for the selection of clinical candidates for B-cell lymphoma indications.


Subject(s)
Lymphoma, B-Cell , Protein-Tyrosine Kinases , Humans , Phospholipase C gamma/metabolism , Signal Transduction , Agammaglobulinaemia Tyrosine Kinase , Lymphoma, B-Cell/drug therapy , Receptors, Antigen, B-Cell/metabolism , Protein Kinase Inhibitors/pharmacology
2.
Sci Signal ; 15(736): eabg5216, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35639855

ABSTRACT

The Bruton's tyrosine kinase (BTK) inhibitor ibrutinib irreversibly binds BTK at Cys481, inhibiting its kinase activity and thus blocking transduction of B cell receptor (BCR) signaling. Although ibrutinib is durably effective in patients with B cell malignancies, many patients still develop ibrutinib-resistant disease. Resistance can arise because of mutations at the ibrutinib-binding site in BTK. Here, we characterized the mechanism by which two BTK mutations, C481F and C481Y, may lead to ibrutinib resistance. Both mutants lacked detectable kinase activity in in vitro kinase assays. Structural modeling suggested that bulky Phe and Tyr side chains at position 481 sterically hinder access to the ATP-binding pocket in BTK, contributing to loss of kinase activity. Nonetheless, BCR signaling still propagated through BTK C481F and C481Y mutants to downstream effectors, the phospholipase PLCγ2 and the transcription factor NF-κB. This maintenance of BCR signaling was partially achieved through the physical recruitment and kinase-independent activation of hematopoietic cell kinase (HCK). Upon BCR activation, BTK C481F or C481Y was phosphorylated by Src family kinases at Tyr551, which then bound to the SH2 domain of HCK. Modeling suggested that this binding disrupted an intramolecular autoinhibitory interaction in HCK. Activated HCK subsequently phosphorylated PLCγ2, which propagated BCR signaling and promoted clonogenic cell proliferation. This kinase-independent mechanism could inform therapeutic approaches to CLL bearing either the C481F or C481Y BTK mutants.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Piperidines , Proto-Oncogene Proteins c-hck , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Drug Resistance, Neoplasm , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Phospholipase C gamma/genetics , Piperidines/pharmacology , Proto-Oncogene Proteins c-hck/metabolism
3.
J Biol Chem ; 289(23): 16270-7, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24719329

ABSTRACT

Compounds that efficiently inhibit the ribonuclease (RNase) H activity of the human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) have yet to be developed. Here, we demonstrate that GSK5750, a 1-hydroxy-pyridopyrimidinone analog, binds to the enzyme with an equilibrium dissociation constant (K(d)) of ~400 nM. Inhibition of HIV-1 RNase H is specific, as DNA synthesis is not affected. Moreover, GSK5750 does not inhibit the activity of Escherichia coli RNase H. Order-of-addition experiments show that GSK5750 binds to the free enzyme in an Mg(2+)-dependent fashion. However, as reported for other active site inhibitors, binding of GSK5750 to a preformed enzyme-substrate complex is severely compromised. The bound nucleic acid prevents access to the RNase H active site, which represents a possible biochemical hurdle in the development of potent RNase H inhibitors. Previous studies suggested that formation of a complex with the prototypic RNase H inhibitor ß-thujaplicinol is slow, and, once formed, it dissociates rapidly. This unfavorable kinetic behavior can limit the potency of RNase H active site inhibitors. Although the association kinetics of GSK5750 remains slow, our data show that this compound forms a long lasting complex with HIV-1 RT. We conclude that slow dissociation of the inhibitor and HIV-1 RT improves RNase H active site inhibitors and may circumvent the obstacle posed by the inability of these compounds to bind to a preformed enzyme-substrate complex.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , Pyridines/pharmacology , Pyrimidinones/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H/antagonists & inhibitors , Base Sequence , Kinetics , Oligodeoxyribonucleotides
4.
PLoS One ; 8(10): e77448, 2013.
Article in English | MEDLINE | ID: mdl-24146996

ABSTRACT

Signature HIV-1 integrase mutations associated with clinical raltegravir resistance involve 1 of 3 primary genetic pathways, Y143C/R, Q148H/K/R and N155H, the latter 2 of which confer cross-resistance to elvitegravir. In accord with clinical findings, in vitro drug resistance profiling studies with wild-type and site-directed integrase mutant viruses have shown significant fold increases in raltegravir and elvitegravir resistance for the specified viral mutants relative to wild-type HIV-1. Dolutegravir, in contrast, has demonstrated clinical efficacy in subjects failing raltegravir therapy due to integrase mutations at Y143, Q148 or N155, which is consistent with its distinct in vitro resistance profile as dolutegravir's antiviral activity against these viral mutants is equivalent to its activity against wild-type HIV-1. Kinetic studies of inhibitor dissociation from wild-type and mutant integrase-viral DNA complexes have shown that dolutegravir also has a distinct off-rate profile with dissociative half-lives substantially longer than those of raltegravir and elvitegravir, suggesting that dolutegravir's prolonged binding may be an important contributing factor to its distinct resistance profile. To provide a structural rationale for these observations, we constructed several molecular models of wild-type and clinically relevant mutant HIV-1 integrase enzymes in complex with viral DNA and dolutegravir, raltegravir or elvitegravir. Here, we discuss our structural models and the posited effects that the integrase mutations and the structural and electronic properties of the integrase inhibitors may have on the catalytic pocket and inhibitor binding and, consequently, on antiviral potency in vitro and in the clinic.


Subject(s)
HIV Integrase Inhibitors/metabolism , HIV Integrase/genetics , HIV-1/genetics , HIV-1/metabolism , Heterocyclic Compounds, 3-Ring/metabolism , Proviruses/genetics , Drug Resistance, Viral/genetics , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV Long Terminal Repeat/genetics , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Kinetics , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Nucleic Acid Conformation , Oxazines , Piperazines , Protein Binding , Protein Conformation , Pyridones
5.
Antimicrob Agents Chemother ; 57(3): 1379-84, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23295935

ABSTRACT

The majority of HIV-1 integrase amino acid sites are highly conserved, suggesting that most are necessary to carry out the critical structural and functional roles of integrase. We analyzed the 34 most variable sites in integrase (>10% variability) and showed that prevalent polymorphic amino acids at these positions did not affect susceptibility to the integrase inhibitor dolutegravir (S/GSK1349572), as demonstrated both in vitro (in site-directed mutagenesis studies) and in vivo (in a phase IIa study of dolutegravir monotherapy in HIV-infected individuals). Ongoing clinical trials will provide additional data on the virologic activity of dolutegravir across subject viruses with and without prevalent polymorphic substitutions.


Subject(s)
HIV Infections/drug therapy , HIV Integrase Inhibitors/therapeutic use , HIV Integrase/genetics , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/therapeutic use , Polymorphism, Genetic , Amino Acid Sequence , Conserved Sequence , HIV Infections/virology , HIV Integrase/metabolism , HIV Integrase Inhibitors/pharmacology , HIV-1/enzymology , HIV-1/genetics , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxazines , Piperazines , Pyridones , Randomized Controlled Trials as Topic
6.
Bioorg Med Chem Lett ; 21(21): 6470-5, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21920742

ABSTRACT

A novel series of pyridyl carboxamide-based CCR5 inhibitors was designed, synthesized, and demonstrated to be highly potent against HIV-1 infection in both HOS and PBL assays. Attempts to evaluate this series of compounds in a rat PK model revealed its instability in rat plasma. A hypothesis for this liability was proposed, and strategies to overcome this issue were pursued, leading to discovery of highly potent 40 and 41, which featured dramatically improved rat PK profiles.


Subject(s)
Anti-HIV Agents/pharmacokinetics , CCR5 Receptor Antagonists , Carboxylic Acids/pharmacokinetics , Amides/chemistry , Animals , Anti-HIV Agents/blood , Anti-HIV Agents/chemistry , Carboxylic Acids/blood , Carboxylic Acids/chemistry , Drug Discovery , Rats
7.
Antimicrob Agents Chemother ; 55(10): 4552-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21807982

ABSTRACT

The integrase inhibitor (INI) dolutegravir (DTG; S/GSK1349572) has significant activity against HIV-1 isolates with raltegravir (RAL)- and elvitegravir (ELV)-associated resistance mutations. As an initial step in characterizing the different resistance profiles of DTG, RAL, and ELV, we determined the dissociation rates of these INIs with integrase (IN)-DNA complexes containing a broad panel of IN proteins, including IN substitutions corresponding to signature RAL and ELV resistance mutations. DTG dissociates slowly from a wild-type IN-DNA complex at 37°C with an off-rate of 2.7 × 10(-6) s(-1) and a dissociative half-life (t(1/2)) of 71 h, significantly longer than the half-lives for RAL (8.8 h) and ELV (2.7 h). Prolonged binding (t(1/2), at least 5 h) was observed for DTG with IN-DNA complexes containing E92, Y143, Q148, and N155 substitutions. The addition of a second substitution to either Q148 or N155 typically resulted in an increase in the off-rate compared to that with the single substitution. For all of the IN substitutions tested, the off-rate of DTG from IN-DNA complexes was significantly slower (from 5 to 40 times slower) than the off-rate of RAL or ELV. These data are consistent with the potential for DTG to have a higher genetic barrier to resistance, provide evidence that the INI off-rate may be an important component of the mechanism of INI resistance, and suggest that the slow dissociation of DTG may contribute to its distinctive resistance profile.


Subject(s)
DNA, Viral/metabolism , HIV Integrase Inhibitors/metabolism , HIV Integrase/metabolism , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/metabolism , Pyrrolidinones/metabolism , Quinolones/metabolism , Amino Acid Substitution , DNA, Complementary , Drug Resistance, Viral , Genotype , HIV Integrase/genetics , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV-1/genetics , Heterocyclic Compounds, 3-Ring/pharmacology , Mutation , Oxazines , Piperazines , Pyridones , Pyrrolidinones/pharmacology , Quinolones/pharmacology , Raltegravir Potassium
9.
Bioorg Med Chem Lett ; 19(2): 469-73, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19056263

ABSTRACT

The evaluation of a series of 4,6-bis-anilino-1H-pyrrolo[2,3-d]pyrimidines as inhibitors of the IGF-1R (IGF-IR) receptor tyrosine kinase is reported. Examples demonstrate nanomolar potencies in in vitro enzyme and mechanistic cellular assays as well as promising in vivo pharmacokinetics in rat.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Drug Discovery , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/chemistry , Rats
10.
Bioorg Med Chem Lett ; 19(2): 373-7, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19081716

ABSTRACT

Initial evaluation of a series 4,6-bis-anilino-1H-pyrrolo[2,3-d]pyrimidines revealed a C(1') carboxamide was preferred for sub-micromolar in vitro potency against IGF-1R. Subsequent solution stability studies with 1 revealed a susceptibility toward acid-induced intramolecular cyclization with the C(1') carboxamide. Herein, we describe several successful approaches toward generating both potent and acid-stable inhibitors of IGF-1R within the 4,6-bis-anilino-1H-pyrrolo[2,3-d]pyrimidine template.


Subject(s)
Acids/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Pyrroles/chemistry
12.
J Chem Inf Model ; 48(12): 2395-403, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19053525

ABSTRACT

The PharmPrint methodology, as modified and implemented by Deanda and Stewart, was prospectively evaluated for use as a virtual high-throughput screening tool by applying it to the design of target-focused arrays. To this end, PharmPrint quantitative structure-activity relationship (QSAR) models for the prediction of AKT1, Aurora-A, and ROCK1 inhibition were constructed and used to virtually screen two large combinatorial libraries. Based on predicted activities, an Aurora-A targeted array and a ROCK1 targeted array were designed and synthesized. One control group per designed array was also synthesized to assess the enrichment levels achieved by the QSAR models. For the Aurora-A targeted array, the hit rate, against the intended target, was 42.9%, whereas that of the control group was 0%. Thus, the enrichment level achieved by the Aurora-A QSAR model was incalculable. For the ROCK1 targeted array, the hit rate against the intended target was 30.6%, whereas that of the control group was 5.10%, making the enrichment level achieved by the ROCK1 QSAR model 6-fold above control. Clearly, these results support the use of the PharmPrint methodology as a virtual screening tool for the design of kinase-targeted arrays.


Subject(s)
Drug Evaluation, Preclinical/statistics & numerical data , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Aurora Kinases , Combinatorial Chemistry Techniques , Computer Simulation , Databases, Protein , Drug Discovery/statistics & numerical data , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quantitative Structure-Activity Relationship , User-Computer Interface , rho-Associated Kinases/antagonists & inhibitors
13.
J Med Chem ; 51(20): 6538-46, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18811134

ABSTRACT

We describe robust chemical approaches toward putative CCR5 scaffolds designed in our laboratories. Evaluation of analogues in the (125)I-[MIP-1beta] binding and Ba-L-HOS antiviral assays resulted in the discovery of 64 and 68 in the 4,4-disubstitited piperidine class H, both potent CCR5 ligands (pIC 50 = 8.30 and 9.00, respectively) and HIV-1 inhibitors (pIC 50 = 7.80 and 7.84, respectively, in Ba-L-HOS assay). In addition, 64 and 68 were bioavailable in rodents, establishing them as lead molecules for further optimization toward CCR5 clinical candidates.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , HIV-1/drug effects , Piperidines/chemical synthesis , Piperidines/pharmacology , Receptors, CCR5/chemistry , Receptors, CCR5/metabolism , Antiviral Agents/chemistry , Drug Evaluation, Preclinical , Ligands , Molecular Structure , Piperidines/chemistry , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 16(20): 5378-83, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16890436

ABSTRACT

The synthesis of several novel aza-stilbene derivatives was carried out. The compounds were tested for their c-RAF enzyme inhibition. Compound 27 possesses significant potency against c-RAF and demonstrates selectivity over other protein kinases. A hypothesis for the binding mode, activity, and selectivity is proposed.


Subject(s)
Aza Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Stilbenes/pharmacology , Aza Compounds/chemical synthesis , Aza Compounds/chemistry , Binding Sites , Enzyme Activation/drug effects , Models, Molecular , Molecular Structure , Protein Conformation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Stereoisomerism , Stilbenes/chemical synthesis , Stilbenes/chemistry , Structure-Activity Relationship
15.
J Chem Inf Comput Sci ; 44(5): 1803-9, 2004.
Article in English | MEDLINE | ID: mdl-15446839

ABSTRACT

The PharmPrint methodology developed by McGregor and Muskal1,2 was used to construct quantitative structure-activity relationship (QSAR) models for the prediction of cyclin-dependent kinase-2 (CDK2) and vascular endothelial growth factor receptor-2 (VEGFR2) inhibition. The QSAR models were constructed based on a binary description of biological activity--a value of zero for inactive and one for active compounds. Subsets of "active" kinase inhibitors (that is, inhibitors with pIC50 > or = 6.0) along with a subset of MDDR3 compounds serving as the recommended set of inactive compounds were used for model development. The predicted activities for the training set compounds were in excellent agreement with the assigned binary activities with greater than 92% of the compounds correctly classified. However, when the QSAR models were applied to the subsets of "inactive" kinase inhibitors (that is, inhibitors with pIC50 < 6.0), greater than 67% were incorrectly predicted to be active. Identical results were obtained with our CDK2 and VEGFR2 validation sets, where the majority of the inactive kinase inhibitors were predicted to be active. In efforts to improve the predictive performance of the QSAR models, simple, but important modifications were made to the PharmPrint methodology. On the basis of these modifications, a second set of QSAR models was constructed and applied to our validation sets to assess their predictive performance. Significant improvements were seen with the modified version of PharmPrint over the original. The results from both versions of PharmPrint are compared and discussed.


Subject(s)
Protein Kinases/chemistry , CDC2-CDC28 Kinases/antagonists & inhibitors , Cyclin-Dependent Kinase 2 , Quantitative Structure-Activity Relationship , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
16.
Mol Pharm ; 1(1): 23-39, 2004 Jan 12.
Article in English | MEDLINE | ID: mdl-15832498

ABSTRACT

A novel, semiempirical approach for the general treatment of solute-solvent interactions (GSSI) was developed to enable the prediction of solution-phase properties (e.g., free energies of desolvation, partition coefficients, and membrane permeabilities). The GSSI approach is based on the principle that all solution-phase processes can be modeled in terms of one or more gas-to-solution transfer processes. The free energy of each gas-to-solution transfer process is calculated as the sum of the free energy of cavity formation and the free energy of solute-solvent interaction. The solute's contributions to these free energies are modeled on the basis of various quantities computed from the solute's three-dimensional (3D) structure, whereas the solvent's contributions are modeled by empirically determined regression coefficients. More specifically, the free energy of cavity formation is modeled on the basis of the total solvent-accessible surface area of the solute. The enthalpy of solute-solvent interaction is modeled on the basis of intermolecular interaction potentials calculated at many uniformly distributed points on the solvent-accessible surface of the solute. The entropy of solute-solvent interaction is modeled on the basis of an effective number of rotatable bonds in the solute and by the regression coefficients characteristic of the solvent. The potential utility of the GSSI approach was demonstrated by modeling the free energy of gas-to-solution transfer for 111 solutes in water, 250 solutes in hexadecane, and 84 solutes in octanol.


Subject(s)
Solutions/chemistry , Solvents/chemistry , Kinetics , Models, Chemical , Thermodynamics
17.
Bioorg Med Chem ; 11(13): 2663-76, 2003 Jul 03.
Article in English | MEDLINE | ID: mdl-12788340

ABSTRACT

This review addresses key pharmacology and virology issues relevant in discovery and development of CCR5 antagonists as anti-HIV drugs, such as target validation, receptor internalization, allosterism, viral resistance and tropism. Recent progress in the discovery and development of CCR5 antagonists, SAR and clinical status are reviewed. Finally, modeling-based structure of CCR5 is discussed in the context of a small-molecule antagonism of the CCR5 receptor.


Subject(s)
Anti-HIV Agents/chemistry , CCR5 Receptor Antagonists , Animals , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/pharmacology , Binding, Competitive , Humans , Ligands , Models, Molecular , Receptors, CCR5/physiology , Structure-Activity Relationship
18.
J Mol Graph Model ; 20(5): 415-25, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11885963

ABSTRACT

A significant number of atoms lie buried beneath the "molecular surface" of proteins and other biologic macromolecules. Interactions between ligands and these macromolecules are dominated by interactions with the "surface atoms". Although interactions with the "buried" or interior atoms of the macromolecule certainly contribute to the total intermolecular interaction energy, many computer-assisted drug design (CADD) strategies can benefit from the identification of those atoms "on the surface" of proteins and other macromolecules. We have developed a simple, yet novel method to distinguish the surface atoms of macromolecules from the interior atoms which is based on computing the atomic contributions to the solvent-accessible surface (SAS) area. This report describes that method and demonstrates that it compares very favorably with four alternative methods.


Subject(s)
Proteins/chemistry , Computer Graphics , Computer Simulation , Computer-Aided Design , Macromolecular Substances , Solvents , Static Electricity , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...