Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 66(13): 8929-8950, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37314941

ABSTRACT

An unmet medical need remains for patients suffering from dry eye disease (DED). A fast-acting, better-tolerated noncorticosteroid anti-inflammatory eye drop could improve patient outcomes and quality of life. Herein, we describe a small-molecule drug discovery effort to identify novel, potent, and water-soluble JAK inhibitors as immunomodulating agents for topical ocular disposition. A focused library of known 3-(4-(2-(arylamino)pyrimidin-4-yl)-1H-pyrazol-1-yl)propanenitriles was evaluated as a molecular starting point. Structure-activity relationships (SARs) revealed a ligand-efficient (LE) JAK inhibitor series, amenable to aqueous solubility. Subsequent in vitro analysis indicated the potential for off-target toxicity. A KINOMEscan selectivity profile of 5 substantiated the likelihood of widespread series affinity across the human kinome. An sp2-to-sp3 drug design strategy was undertaken to attenuate off-target kinase activity while driving JAK-STAT potency and aqueous solubility. Tactics to reduce aromatic character, increase fraction sp3 (Fsp3), and bolster molecular complexity led to the azetidin-3-amino bridging scaffold in 31.


Subject(s)
Janus Kinase Inhibitors , Humans , Janus Kinase 1 , Janus Kinase 2 , Janus Kinase 3 , Janus Kinase Inhibitors/pharmacology , Janus Kinases , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Solubility
2.
J Ocul Pharmacol Ther ; 39(2): 117-127, 2023 03.
Article in English | MEDLINE | ID: mdl-36602977

ABSTRACT

Purpose: A new class of ocular steroids designed to mitigate steroid-induced intraocular pressure (IOP) elevation while maintaining anti-inflammatory activity was developed. Herein is described the discovery and preclinical characterization of ROCK'Ster compound 1. Methods: Codrugs consisting of a Rho kinase inhibitor (ROCKi) and a corticosteroid were synthesized. Compounds were initially screened in vitro for ROCKi activity and anti-inflammatory activity against the proinflammatory interleukin 23 and bacterial lipopolysaccharide (LPS) pathways. Selected compounds were then screened for solubility, chemical stability, and ex vivo corneal metabolism. Lead compound 1 was evaluated for IOP lowering in the Dutch Belted rabbit and for anti-inflammatory efficacy in both a postcataract surgery model and an allergic eye disease (AED) mouse model. Results: Several ROCK'Sters were found to be potent inhibitors of ROCK (Kis < 50 nM), have high anti-inflammatory activity in vitro (IC50s < 50 nM), display sufficient stability in topical ophthalmic formulations, and have a moderate rate of corneal metabolism. Compound 1 (0.1% and 0.25%, quater in die [QID]-4 times a day) demonstrated IOP-lowering capability without inducing hyperemia in our rabbit model. When compared with the marketed steroids, Durezol® and Pred Forte®, compound 1 (0.1%, 0.25%) demonstrated noninferiority in clinical scoring in a rabbit model of inflammation after surgery. In addition, anti-inflammatory outcomes were observed with compound 1 (0.1%) relative to Lotemax® or vehicle control in an AED mouse model. Conclusion: ROCK'Ster compound 1 is a novel compound suitable for topical ocular dosing that possesses IOP-lowering capability along with similar anti-inflammatory activity compared with marketed steroids.


Subject(s)
Eye Diseases , Intraocular Pressure , Animals , Mice , Rabbits , rho-Associated Kinases , Eye Diseases/drug therapy , Disease Models, Animal , Adrenal Cortex Hormones , Cornea , Ophthalmic Solutions/pharmacology , Antihypertensive Agents
3.
Bioorg Med Chem ; 28(23): 115791, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33059303

ABSTRACT

GlaxoSmithKline and Astex Pharmaceuticals recently disclosed the discovery of the potent H-PGDS inhibitor GSK2894631A 1a (IC50 = 9.9 nM) as part of a fragment-based drug discovery collaboration with Astex Pharmaceuticals. This molecule exhibited good murine pharmacokinetics, allowing it to be utilized to explore H-PGDS pharmacology in vivo. Yet, with prolonged dosing at higher concentrations, 1a induced CNS toxicity. Looking to attenuate brain penetration in this series, aza-quinolines, were prepared with the intent of increasing polar surface area. Nitrogen substitutions at the 6- and 8-positions of the quinoline were discovered to be tolerated by the enzyme. Subsequent structure activity studies in these aza-quinoline scaffolds led to the identification of 1,8-naphthyridine 1y (IC50 = 9.4 nM) as a potent peripherally restricted H-PGDS inhibitor. Compound 1y is efficacious in four in vivo inflammatory models and exhibits no CNS toxicity.


Subject(s)
Aza Compounds/chemistry , Enzyme Inhibitors/chemistry , Quinolines/chemistry , Animals , Binding Sites , Brain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Drug Stability , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Kinetics , Male , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Rats , Structure-Activity Relationship
4.
Skelet Muscle ; 10(1): 30, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33092650

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disorder stemming from a loss of functional dystrophin. Current therapeutic options for DMD are limited, as small molecule modalities remain largely unable to decrease the incidence or mitigate the consequences of repetitive mechanical insults to the muscle during eccentric contractions (ECCs). METHODS: Using a metabolomics-based approach, we observed distinct and transient molecular phenotypes in muscles of dystrophin-deficient MDX mice subjected to ECCs. Among the most chronically depleted metabolites was nicotinamide adenine dinucleotide (NAD), an essential metabolic cofactor suggested to protect muscle from structural and metabolic degeneration over time. We tested whether the MDX muscle NAD pool can be expanded for therapeutic benefit using two complementary small molecule strategies: provision of a biosynthetic precursor, nicotinamide riboside, or specific inhibition of the NAD-degrading ADP-ribosyl cyclase, CD38. RESULTS: Administering a novel, potent, and orally available CD38 antagonist to MDX mice successfully reverted a majority of the muscle metabolome toward the wildtype state, with a pronounced impact on intermediates of the pentose phosphate pathway, while supplementing nicotinamide riboside did not significantly affect the molecular phenotype of the muscle. However, neither strategy sustainably increased the bulk tissue NAD pool, lessened muscle damage markers, nor improved maximal hindlimb strength following repeated rounds of eccentric challenge and recovery. CONCLUSIONS: In the absence of dystrophin, eccentric injury contributes to chronic intramuscular NAD depletion with broad pleiotropic effects on the molecular phenotype of the tissue. These molecular consequences can be more effectively overcome by inhibiting the enzymatic activity of CD38 than by supplementing nicotinamide riboside. However, we found no evidence that either small molecule strategy is sufficient to restore muscle contractile function or confer protection from eccentric injury, undermining the modulation of NAD metabolism as a therapeutic approach for DMD.


Subject(s)
Enzyme Inhibitors/pharmacology , Metabolome , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/drug therapy , NAD/metabolism , Niacinamide/analogs & derivatives , Pyridinium Compounds/pharmacology , ADP-ribosyl Cyclase 1/antagonists & inhibitors , Animals , Dystrophin/deficiency , Enzyme Inhibitors/therapeutic use , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Pyridinium Compounds/therapeutic use
5.
Nat Commun ; 10(1): 4659, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604915

ABSTRACT

MG53 is a muscle-specific TRIM-family protein that presides over the cell membrane repair response. Here, we show that MG53 present in blood circulation acts as a myokine to facilitate tissue injury-repair and regeneration. Transgenic mice with sustained elevation of MG53 in the bloodstream (tPA-MG53) have a healthier and longer life-span when compared with littermate wild type mice. The tPA-MG53 mice show normal glucose handling and insulin signaling in skeletal muscle, and sustained elevation of MG53 in the bloodstream does not have a deleterious impact on db/db mice. More importantly, the tPA-MG53 mice display remarkable dermal wound healing capacity, enhanced muscle performance, and improved injury-repair and regeneration. Recombinant human MG53 protein protects against eccentric contraction-induced acute and chronic muscle injury in mice. Our findings highlight the myokine function of MG53 in tissue protection and present MG53 as an attractive biological reagent for regenerative medicine without interference with glucose handling in the body.


Subject(s)
Membrane Proteins/physiology , Wound Healing , Animals , Calcium/metabolism , Glucose/metabolism , Glucose Tolerance Test , Insulin/metabolism , Membrane Proteins/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Regeneration/genetics , Systems Biology
6.
Skelet Muscle ; 8(1): 38, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30526662

ABSTRACT

Following publication of the original article [1], the authors flagged that there is a discrepancy with the Availability of data and materials statement on page 12 of the article.

7.
Skelet Muscle ; 8(1): 35, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30424786

ABSTRACT

BACKGROUND: In muscular dystrophy and old age, skeletal muscle repair is compromised leading to fibrosis and fatty tissue accumulation. Therefore, therapies that protect skeletal muscle or enhance repair would be valuable medical treatments. Hypoxia-inducible factors (HIFs) regulate gene transcription under conditions of low oxygen, and HIF target genes EPO and VEGF have been associated with muscle protection and repair. We tested the importance of HIF activation following skeletal muscle injury, in both a murine model and human volunteers, using prolyl hydroxylase inhibitors that stabilize and activate HIF. METHODS: Using a mouse eccentric limb injury model, we characterized the protective effects of prolyl hydroxylase inhibitor, GSK1120360A. We then extended these studies to examine the impact of EPO modulation and infiltrating immune cell populations on muscle protection. Finally, we extended this study with an experimental medicine approach using eccentric arm exercise in untrained volunteers to measure the muscle-protective effects of a clinical prolyl hydroxylase inhibitor, daprodustat. RESULTS: GSK1120360A dramatically prevented functional deficits and histological damage, while accelerating recovery after eccentric limb injury in mice. Surprisingly, this effect was independent of EPO, but required myeloid HIF1α-mediated iNOS activity. Treatment of healthy human volunteers with high-dose daprodustat reduced accumulation of circulating damage markers following eccentric arm exercise, although we did not observe any diminution of functional deficits with compound treatment. CONCLUSION: The results of these experiments highlight a novel skeletal muscle protective effect of prolyl hydroxylase inhibition via HIF-mediated expression of iNOS in macrophages. Partial recapitulation of these findings in healthy volunteers suggests elements of consistent pharmacology compared to responses in mice although there are clear differences between these two systems.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glycine/analogs & derivatives , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Muscle Contraction , Muscle, Skeletal/drug effects , Myalgia/drug therapy , Quinolones/therapeutic use , Adolescent , Adult , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glycine/pharmacology , Glycine/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Myalgia/etiology , Quinolones/pharmacology
8.
ACS Chem Biol ; 11(2): 518-29, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26696218

ABSTRACT

Skeletal muscle progenitor stem cells (referred to as satellite cells) represent the primary pool of stem cells in adult skeletal muscle responsible for the generation of new skeletal muscle in response to injury. Satellite cells derived from aged muscle display a significant reduction in regenerative capacity to form functional muscle. This decrease in functional recovery has been attributed to a decrease in proliferative capacity of satellite cells. Hence, agents that enhance the proliferative abilities of satellite cells may hold promise as therapies for a variety of pathological settings, including repair of injured muscle and age- or disease-associated muscle wasting. Through phenotypic screening of isolated murine satellite cells, we identified a series of 2,4-diaminopyrimidines (e.g., 2) that increased satellite cell proliferation. Importantly, compound 2 was effective in accelerating repair of damaged skeletal muscle in an in vivo mouse model of skeletal muscle injury. While these compounds were originally prepared as c-Jun N-terminal kinase 1 (JNK-1) inhibitors, structure-activity analyses indicated JNK-1 inhibition does not correlate with satellite cell activity. Screening against a broad panel of kinases did not result in identification of an obvious molecular target, so we conducted cell-based proteomics experiments in an attempt to identify the molecular target(s) responsible for the potentiation of the satellite cell proliferation. These data provide the foundation for future efforts to design improved small molecules as potential therapeutics for muscle repair and regeneration.


Subject(s)
Cell Proliferation/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Regeneration/drug effects , 3T3 Cells , Animals , Cells, Cultured , Drug Discovery , Humans , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/injuries , Pyrimidines/pharmacokinetics , Stem Cells/cytology , Stem Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...