Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Med Chem Lett ; 12(11): 1847-1852, 2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34795875

ABSTRACT

Human ß-nerve growth factor (ß-NGF) and its associated receptor, human tropomyosin receptor kinase A (hTrkA), have been demonstrated to be key factors in the perception of pain. However, efficacious small molecule therapies targeting the intracellularly located hTrkA kinase have not been explored thoroughly for pain management. Herein, we report the pharmacological properties of a selective hTrkA allosteric inhibitor, 1. 1 was shown to be active against the full length hTrkA, showing preferential binding for the inactive kinase, and was confirmed through the X-ray of hTrkA···1 bound complex. 1 was also found to inhibit ß-NGF induced neurite outgrowth in rat PC12 cells. Daily oral administration of 1 improved the joint compression threshold of rats injected intra-articularly with monoiodoacetate over a 14-day period. The efficacy of 1 in a relevant chronic pain model of osteoarthritis coupled with in vitro confirmation of target mediation makes allosteric hTrkA inhibitors potential candidates for modulating pain.

2.
RSC Med Chem ; 11(3): 370-377, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-33479642

ABSTRACT

In silico virtual screening followed by in vitro biochemical, biophysical, and cellular screening resulted in the identification of distinctly different hTrkA kinase domain inhibitor scaffolds. X-ray structural analysis of representative inhibitors bound to hTrkA kinase domain defined the binding mode and rationalized the mechanism of action. Preliminary assessment of the sub-type selectivity against the closest hTrkB isoform, and early ADME guided the progression of select inhibitor leads in the screening cascade. The possibility of the actives sustaining to known hTrkA resistance mutations assessed in silico offers initial guidance into the required multiparametric lead optimization to arrive at a clinical candidate.

3.
Bioorg Med Chem Lett ; 29(22): 126680, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31610943

ABSTRACT

Virtual in silico structure-guided modeling, followed by in vitro biochemical screening of a subset of commercially purchasable compound collection resulted in the identification of several human tropomyosin receptor kinase A (hTrkA) inhibitors that bind the orthosteric ATP site and exhibit binding preference for the inactive kinase conformation. The type 2 binding mode with the DFG-out and αC-helix out hTrkA kinase domain conformation was confirmed from X-ray crystallographic solution of a representative inhibitor analog, 1b. Additional hTrkA and hTrkB (selectivity) assays in recombinant cells, neurite outgrowth inhibition using rat PC12 cells, early ADME profiling, and preliminary pharmacokinetic evaluation in rodents guided the lead inhibitor progression in the discovery screening funnel.


Subject(s)
Receptor, trkA/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Neuronal Outgrowth/drug effects , PC12 Cells , Rats , Receptor, trkA/metabolism , Structure-Activity Relationship
4.
Bioorg Med Chem Lett ; 29(19): 126624, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31444087

ABSTRACT

In silico virtual screening using the ligand-based ROCS approach and the commercially purchasable compound collection from the ZINC database resulted in the identification of distinctly different and novel acetamide core frameworks with series representatives 1a and 2a exhibiting nanomolar affinity in the kinase domain only hTrkA HTRF biochemical assay. Additional experimental validation using the Caliper technology with either the active or inactive kinase conditions demonstrated the leads, 1a and 2a, to preferentially bind the kinase inactive state. X-ray structural analysis of the kinase domain of hTrkA…1a/2a complexes confirmed the kinase, bind the inhibitor leads in the inactive state and to exhibit a type 2 binding mode with the DFG-out and αC-helix out conformation. The leads also demonstrated sub-micromolar activity in the full length hTrkA cell-based assay and selectivity against the closely related hTrkB isoform. However, the poor microsomal stability and permeability of the leads is suggestive of a multiparametric lead optimization effort requirement for further progression.


Subject(s)
Drug Design , Protein Kinase Inhibitors/pharmacology , Receptor, trkA/antagonists & inhibitors , Computer Simulation , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Receptor, trkA/chemistry , Structure-Activity Relationship
5.
ACS Chem Biol ; 14(6): 1205-1216, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31059222

ABSTRACT

Access to cryptic binding pockets or allosteric sites on a kinase that present themselves when the enzyme is in a specific conformational state offers a paradigm shift in designing the next generation small molecule kinase inhibitors. The current work showcases an extensive and exhaustive array of in vitro biochemical and biophysical tools and techniques deployed along with structural biology efforts of inhibitor-bound kinase complexes to characterize and confirm the cryptic allosteric binding pocket and docking mode of the small molecule actives identified for hTrkA. Specifically, assays were designed and implemented to lock the kinase in a predominantly active or inactive conformation and the effect of the kinase inhibitor probed to understand the hTrkA binding and hTrkB selectivity. The current outcome suggests that inhibitors with a fast association rate take advantage of the inactive protein conformation and lock the kinase state by also exhibiting a slow off-rate. This in turn shifts the inactive/active state protein conformational equilibrium cycle, affecting the subsequent downstream signaling.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptor, trkA/antagonists & inhibitors , Allosteric Regulation , Animals , Computer Simulation , Humans , Ligands , Neurites , PC12 Cells , Protein Kinase Inhibitors/metabolism , Rats , Receptor, trkA/metabolism
6.
Biotechnol Bioeng ; 115(8): 1925-1937, 2018 08.
Article in English | MEDLINE | ID: mdl-29663315

ABSTRACT

Nerve growth factor (NGF) plays a central role in multiple chronic pain conditions. As such, anti-NGF monoclonal antibodies (mAbs) that function by antagonizing NGF downstream signaling are leading drug candidates for non-opioid pain relief. To evaluate anti-canine NGF (cNGF) mAbs we sought a yeast surface display platform of cNGF. Both mature cNGF and pro-cNGF displayed on the yeast surface but bound conformationally sensitive mAbs at most 2.5-fold in mean fluorescence intensity above background, suggesting that cNGF was mostly misfolded. To improve the amount of folded, displayed cNGF, we used comprehensive mutagenesis, FACS, and deep sequencing to identify point mutants in the pro-region of canine NGF that properly enhance the folded protein displayed on the yeast surface. Out of 1,737 tested single point mutants in the pro region, 49 increased the amount of NGF recognized by conformationally sensitive mAbs. These gain-of-function mutations cluster around residues A-61-P-26. Gain-of-function mutants were additive, and a construct containing three mutations increased amount of folded cNGF to 23-fold above background. Using this new cNGF construct, fine conformational epitopes for tanezumab and three anti-cNGF mAbs were evaluated. The epitope revealed by the yeast experiments largely overlapped with the tanezumab epitope previously determined by X-ray crystallography. The other mAbs showed site-specific differences with tanezumab. As the number of binding epitopes of functionally neutralizing anti-NGF mAbs on NGF are limited, subtle differences in the individual interacting residues on NGF that bind each mAb contribute to the understanding of each antibody and variations in its neutralizing activity. These results demonstrate the potential of deep sequencing-guided protein engineering to improve the production of folded surface-displayed protein, and the resulting cNGF construct provides a platform to map conformational epitopes for other anti-neurotrophin mAbs.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cell Surface Display Techniques/methods , Epitope Mapping , Mutant Proteins/metabolism , Nerve Growth Factor/metabolism , Yeasts/metabolism , Mutant Proteins/genetics , Nerve Growth Factor/genetics , Protein Binding , Yeasts/genetics
7.
Bioorg Med Chem Lett ; 21(21): 6545-53, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21924613

ABSTRACT

Fatty acid amide hydrolase (FAAH) is an integral membrane serine hydrolase responsible for the degradation of fatty acid amide signaling molecules such as endocannabinoid anandamide (AEA), which has been shown to possess cannabinoid-like analgesic properties. Herein we report the optimization of spirocyclic 7-azaspiro[3.5]nonane and 1-oxa-8-azaspiro[4.5]decane urea covalent inhibitors of FAAH. Using an iterative design and optimization strategy, lead compounds were identified with a remarkable reduction in molecular weight and favorable CNS drug like properties. 3,4-Dimethylisoxazole and 1-methyltetrazole were identified as superior urea moieties for this inhibitor class. A dual purpose in vivo efficacy and pharmacokinetic screen was designed to be the key decision enabling experiment affording the ability to move quickly from compound synthesis to selection of preclinical candidates. On the basis of the remarkable potency, selectivity, pharmacokinetic properties and in vivo efficacy, PF-04862853 (15p) was advanced as a clinical candidate.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Pain/drug therapy , Spiro Compounds/pharmacology , Administration, Oral , Analgesics/administration & dosage , Analgesics/chemistry , Analgesics/therapeutic use , Animals , Aza Compounds/administration & dosage , Aza Compounds/chemistry , Aza Compounds/pharmacology , Aza Compounds/therapeutic use , Biological Availability , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Rats , Spiro Compounds/administration & dosage , Spiro Compounds/chemistry , Spiro Compounds/therapeutic use
8.
Bioorg Med Chem Lett ; 21(21): 6538-44, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21924614

ABSTRACT

Herein we report the identification of two new fatty acid amide hydrolase (FAAH) inhibitor lead series with FAAH k(inact)/K(i) potency values greater than 1500M(-1)s(-1). The two novel spirocyclic cores, 7-azaspiro[3.5]nonane and 1-oxa-8-azaspiro[4.5]decane, clearly distinguished themselves from the other spirocyclic cores on the basis of their superior potency for FAAH. Lead compounds from these two series have suitable FAAH potency and selectivity for additional medicinal chemistry optimization.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Aza Compounds/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Spiro Compounds/pharmacology , Aza Compounds/chemistry , Enzyme Inhibitors/chemistry , Models, Molecular , Spiro Compounds/chemistry , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 19(20): 5970-4, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19765986

ABSTRACT

Fatty acid amide hydrolase (FAAH) has attracted significant attention due to its promise as an analgesic target. This has resulted in the discovery of numerous chemical classes as inhibitors of this potential therapeutic target. In this paper we disclose a new series of novel FAAH irreversible azetidine urea inhibitors. In general these compounds illustrate potent activity against the rat FAAH enzyme. Our SAR studies allowed us to optimize this series resulting in the identification of compounds 13 which were potent inhibitors of both human and rat enzyme. This series of compounds illustrated good hydrolase selectivity along with good PK properties.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/chemistry , Azetidines/chemistry , Enzyme Inhibitors/chemistry , Pyridazines/chemistry , Urea/chemistry , Amidohydrolases/metabolism , Analgesics/chemical synthesis , Analgesics/pharmacokinetics , Animals , Azetidines/chemical synthesis , Azetidines/pharmacokinetics , Binding Sites , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Humans , Pyridazines/chemical synthesis , Pyridazines/pharmacokinetics , Rats , Structure-Activity Relationship , Urea/chemical synthesis , Urea/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...