Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 14(2): 191-198, 2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36793423

ABSTRACT

Macrocyclic retinoic acid receptor-related orphan receptor C2 (RORC2) inverse agonists have been designed with favorable properties for topical administration. Inspired by the unanticipated bound conformation of an acyclic sulfonamide-based RORC2 ligand from cocrystal structure analysis, macrocyclic linker connections between the halves of the molecule were explored. Further optimization of analogues was accomplished to maximize potency and refine physiochemical properties (MW, lipophilicity) best suited for topical application. Compound 14 demonstrated potent inhibition of interleukin-17A (IL-17A) production by human Th17 cells and in vitro permeation through healthy human skin achieving high total compound concentration in both skin epidermis and dermis layers.

2.
Br J Clin Pharmacol ; 84(8): 1776-1788, 2018 08.
Article in English | MEDLINE | ID: mdl-29672897

ABSTRACT

AIMS: To determine the safety, tolerability, pharmacokinetics and pharmacodynamics of the Janus kinase 1-selective inhibitor, PF-04965842. METHODS: This was a phase 1, first-in-human, randomized, double-blind, placebo-controlled, combination single- and multiple-dose escalation, parallel design study in healthy subjects (http://clinicaltrials.gov, NCT01835197). Subjects received a single dose of placebo or 3, 10, 30, 100, 200, 400 or 800 mg PF-04965842 (single ascending dose phase) and placebo or 30 mg once daily (QD), 100 mg QD, 200 mg QD, 400 mg QD, 100 mg twice daily (BID) or 200 mg BID PF-04965842 for 10 consecutive days (multiple ascending dose phase). The primary objective was to determine the safety and tolerability of PF-04965842. RESULTS: Seventy-nine subjects were randomized and received study treatments. There were no deaths or serious adverse events. The most frequent treatment-emergent adverse events were headache (n = 13), diarrhoea (n = 11) and nausea (n = 11). PF-04965842 was absorbed rapidly (median time at which maximum plasma concentration occurred generally ≤1 h following either single- or multiple-dose administration) and eliminated rapidly (mean t½ 2.8-5.2 h after 10 days of QD or BID administration in the multiple ascending dose phase). Increases in maximum plasma concentration and area under the concentration-time curve were dose proportional up to 200 mg (single or total daily doses) with an apparent trend towards greater than proportional increases with higher doses. Less than 4.4% of the dose was recovered unchanged in urine. Changes in pharmacodynamic biomarkers were consistent with the known effects of Janus kinase signalling inhibition. CONCLUSIONS: These results support further evaluation of PF-04965842 for clinical use in patients with inflammatory diseases.


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Adult , Area Under Curve , Diarrhea/chemically induced , Diarrhea/epidemiology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Headache/chemically induced , Headache/epidemiology , Humans , Male , Middle Aged , Nausea/chemically induced , Nausea/epidemiology , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Young Adult
3.
J Med Chem ; 61(3): 1130-1152, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29298069

ABSTRACT

Janus kinases (JAKs) are intracellular tyrosine kinases that mediate the signaling of numerous cytokines and growth factors involved in the regulation of immunity, inflammation, and hematopoiesis. As JAK1 pairs with JAK2, JAK3, and TYK2, a JAK1-selective inhibitor would be expected to inhibit many cytokines involved in inflammation and immune function while avoiding inhibition of the JAK2 homodimer regulating erythropoietin and thrombopoietin signaling. Our efforts began with tofacitinib, an oral JAK inhibitor approved for the treatment of rheumatoid arthritis. Through modification of the 3-aminopiperidine linker in tofacitinib, we discovered highly selective JAK1 inhibitors with nanomolar potency in a human whole blood assay. Improvements in JAK1 potency and selectivity were achieved via structural modifications suggested by X-ray crystallographic analysis. After demonstrating efficacy in a rat adjuvant-induced arthritis (rAIA) model, PF-04965842 (25) was nominated as a clinical candidate for the treatment of JAK1-mediated autoimmune diseases.


Subject(s)
Autoimmune Diseases/drug therapy , Cyclobutanes/pharmacology , Janus Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Sulfonamides/pharmacology , Animals , Arthritis, Experimental/drug therapy , Cyclobutanes/chemistry , Cyclobutanes/pharmacokinetics , Cyclobutanes/therapeutic use , Dogs , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Janus Kinase 1/chemistry , Janus Kinase 2/antagonists & inhibitors , Models, Molecular , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use , Rats , Substrate Specificity , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Tissue Distribution
4.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 11): 840-845, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27827355

ABSTRACT

Crystals of phosphorylated JAK1 kinase domain were initially generated in complex with nucleotide (ADP) and magnesium. The tightly bound Mg2+-ADP at the ATP-binding site proved recalcitrant to ligand displacement. Addition of a molar excess of EDTA helped to dislodge the divalent metal ion, promoting the release of ADP and allowing facile exchange with ATP-competitive small-molecule ligands. Many kinases require the presence of a stabilizing ligand in the ATP site for crystallization. This procedure could be useful for developing co-crystallization systems with an exchangeable ligand to enable structure-based drug design of other protein kinases.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Crystallization/methods , Edetic Acid/chemistry , Janus Kinase 1/chemistry , Magnesium/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Binding Sites , Cations, Divalent , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Magnesium/metabolism , Models, Molecular , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera
5.
Am J Physiol Renal Physiol ; 306(1): F68-74, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24197070

ABSTRACT

The importance of membrane-bound PGE synthase 1 (mPGES1) in the regulation of renal function has been examined in mPGES1-deficient mice or by evaluating changes in its expression. However, it is unknown whether prolonged mPGES1 inhibition induces significant changes of renal function when Na(+) intake is normal or low. This study examined the renal effects elicited by a selective mPGES1 inhibitor (PF-458) during 7 days in conscious chronically instrumented dogs with normal Na(+) intake (NSI) or low Na(+) intake (LSI). Results obtained in both in vitro and in vivo studies have strongly suggested that PF-458 is a selective mPGES1 inhibitor. The administration of 2.4 mg·kg(-1)·day(-1) PF-458 to dogs with LSI did not induce significant changes in renal blood flow (RBF) and glomerular filtration rate (GFR). A larger dose of PF-458 (9.6 mg·kg(-1)·day(-1)) reduced RBF (P < 0.05) but not GFR in dogs with LSI and did not induce changes of renal hemodynamic in dogs with NSI. Both doses of PF-458 elicited a decrease (P < 0.05) in PGE2 and an increase (P < 0.05) in 6-keto-PGF1α. The administration of PF-458 did not induce significant changes in renal excretory function, plasma renin activity, and plasma aldosterone and thromboxane B2 concentrations in dogs with LSI or NSI. The results obtained suggest that mPGES1 is involved in the regulation of RBF when Na(+) intake is low and that the renal effects elicited by mPGES1 inhibition are modulated by a compensatory increment in PGI2. These results may have some therapeutical implications since it has been shown that prolonged mPGES1 inhibition has lower renal effects than those elicited by nonsteroidal anti-inflammatory drugs or selective cyclooxygenase-2 inhibitors.


Subject(s)
Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Kidney/physiology , Renal Circulation/physiology , Sodium/pharmacology , Aldosterone/blood , Animals , Benzoxazoles/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Dogs , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Intramolecular Oxidoreductases/genetics , Kidney/drug effects , Piperidines/pharmacology , Potassium/urine , Prostaglandin-E Synthases , Renal Circulation/drug effects , Sodium/administration & dosage , Sodium/urine , Thromboxane B2/blood
6.
Bioorg Med Chem Lett ; 23(4): 1120-6, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23298810

ABSTRACT

Microsomal prostaglandin E(2) synthase-1 (mPGES-1) is a novel therapeutic target for the treatment of inflammation and pain. In the preceding letter, we detailed the discovery of clinical candidate PF-04693627, a potent mPGES-1 inhibitor possessing a novel benzoxazole structure. While PF-04693627 was undergoing further preclinical profiling, we sought to identify a back-up mPGES-1 inhibitor that differentiated itself from PF-04693627. The design, synthesis, mPGES-1 activity and in vivo PK of a novel set of substituted benzoxazoles are described herein. Also described is a conformation-based hypothesis for mPGES-1 activity based on the preferred conformation of the cyclohexane ring within this class of inhibitors.


Subject(s)
Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Benzoxazoles/chemical synthesis , Drug Design , Enzyme Inhibitors/chemical synthesis , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/metabolism , Models, Molecular , Molecular Conformation , Prostaglandin-E Synthases , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 23(4): 1114-9, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23260349

ABSTRACT

Inhibition of mPGES-1, the terminal enzyme in the arachidonic acid/COX pathway to regulate the production of pro-inflammatory prostaglandin PGE2, is considered an attractive new therapeutic target for safe and effective anti-inflammatory drugs. The discovery of a novel series of orally active, selective benzoxazole piperidinecarboxamides as mPGES-1 inhibitors is described. Structure-activity optimization of lead 5 with cyclohexyl carbinols resulted in compound 12, which showed excellent in vitro potency and selectivity against COX-2, and reasonable pharmacokinetic properties. Further SAR studies of the benzoxazole ring substituents lead to a novel series of highly potent compounds with improved PK profile, including 23, 26, and 29, which were effective in a carrageenan-stimulated guinea pig air pouch model of inflammation. Based on its excellent in vitro and in vivo pharmacological, pharmacokinetic and safety profile and ease of synthesis, compound 26 (PF-4693627) was advanced to clinical studies.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Inflammation/drug therapy , Intramolecular Oxidoreductases/antagonists & inhibitors , Drug Discovery , Humans , Inflammation/enzymology , Intramolecular Oxidoreductases/metabolism , Prostaglandin-E Synthases , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 23(3): 907-11, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23266122

ABSTRACT

A novel series of potent benzoxazole mPGES-1 inhibitors has been derived from a hit from a high throughput screen. Compound 37 displays mPGES-1 inhibition in an enzyme assay (0.018 µM) and PGE-2 inhibition in a cell-based assay (0.034 µM). It demonstrates 500- and 2500-fold selectivity for mPGES-1 over COX-2 and 6-keto PGF-1α, respectively. In vivo PK studies in dogs demonstrate 55% oral bioavailability and an 7 h half-life.


Subject(s)
Benzoxazoles/chemistry , Enzyme Inhibitors/chemistry , Intramolecular Oxidoreductases/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Amides/pharmacology , Animals , Benzoxazoles/chemical synthesis , Benzoxazoles/pharmacokinetics , Benzoxazoles/pharmacology , Biological Availability , Dogs , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Molecular Structure , Prostaglandin-E Synthases , Structure-Activity Relationship
9.
Biochem Pharmacol ; 79(10): 1445-54, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20067770

ABSTRACT

Inflammation-induced microsomal prostaglandin E synthase-1 (mPGES-1) is the terminal enzyme that synthesizes prostaglandin E(2) (PGE(2)) downstream of cyclooxygenase-2 (COX-2). The efficacy of nonsteroidal anti-inflammatory drugs and COX-2 inhibitors in the treatment of the signs and symptoms of osteoarthritis, rheumatoid arthritis and inflammatory pain, largely attributed to the inhibition of PGE(2) synthesis, provides a rationale for exploring mPGES-1 inhibition as a potential novel therapy for these diseases. Toward this aim, we identified PF-9184 as a novel mPGES-1 inhibitor. PF-9184 potently inhibited recombinant human (rh) mPGES-1 (IC(50)=16.5+/-3.8nM), and had no effect against rhCOX-1 and rhCOX-2 (>6500-fold selectivity). In inflammation and clinically relevant biological systems, mPGES-1 expression, like COX-2 expression was induced in cell context- and time-dependent manner, consistent with the kinetics of PGE(2) synthesis. In rationally designed cell systems ideal for determining direct effects of the inhibitors on mPGES-1 function, but not its expression, PF-9184 inhibited PGE(2) synthesis (IC(50) in the range of 0.5-5 microM in serum-free cell and human whole blood cultures, respectively) while sparing the synthesis of 6-keto-PGF(1alpha) (PGF(1alpha)) and PGF(2alpha). In contrast, as expected, the selective COX-2 inhibitor, SC-236, inhibited PGE(2), PGF(1alpha) and PGF(2alpha) synthesis. This profile of mPGES-1 inhibition, distinct from COX-2 inhibition in cells, validates mPGES-1 as an attractive target for therapeutic intervention.


Subject(s)
Cyclic S-Oxides/antagonists & inhibitors , Cyclooxygenase 2 Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Thiazines/antagonists & inhibitors , Animals , Arthritis, Rheumatoid/metabolism , Carrageenan/pharmacology , Cells, Cultured , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression/drug effects , Humans , Immunoblotting , Interleukin-1beta/pharmacology , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/metabolism , Microsomes/drug effects , Microsomes/enzymology , Prostaglandin-E Synthases , Rats , Reverse Transcriptase Polymerase Chain Reaction
10.
J Comput Aided Mol Des ; 23(1): 13-24, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18777160

ABSTRACT

Inducible, microsomal prostaglandin E synthase 1 (mPGES-1), the terminal enzyme in the prostaglandin (PG) biosynthetic pathway, constitutes a promising therapeutic target for the development of new anti-inflammatory drugs. To elucidate structure-function relationships and to enable structure-based design, an mPGES-1 homology model was developed using the three-dimensional structure of the closest homologue of the MAPEG family (Membrane Associated Proteins in Eicosanoid and Glutathione metabolism), mGST-1. The ensuing model of mPGES-1 is a homo-trimer, with each monomer consisting of four membrane-spanning segments. Extensive structure refinement revealed an inter-monomer salt bridge (K26-E77) as well as inter-helical interactions within each monomer, including polar hydrogen bonds (e.g. T78-R110-T129) and hydrophobic pi-stacking (F82-F103-F106), all contributing to the overall stability of the homo-trimer of mPGES-1. Catalytic co-factor glutathione (GSH) was docked into the mPGES-1 model by flexible optimization of both the ligand and the protein conformations, starting from the initial location ascertained from the mGST-1 structure. Possible binding site for the substrate, prostaglandin H(2) (PGH(2)), was identified by systematically probing the refined molecular structure of mPGES-1. A binding model was generated by induced fit docking of PGH(2) in the presence of GSH. The homology model prescribes three potential inhibitor binding sites per mPGES-1 trimer. This was further confirmed experimentally by equilibrium dialysis study which generated a binding stoichiometric ratio of approximately three inhibitor molecules to three mPGES-1 monomers. The structural model that we have derived could serve as a useful tool for structure-guided design of inhibitors for this emergently important therapeutic target.


Subject(s)
Enzyme Inhibitors/chemistry , Intramolecular Oxidoreductases/chemistry , Microsomes/enzymology , Amino Acid Sequence , Biopolymers , Enzyme Inhibitors/pharmacology , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Models, Molecular , Molecular Sequence Data , Prostaglandin-E Synthases , Protein Conformation , Sequence Homology, Amino Acid
11.
J Med Chem ; 50(23): 5712-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17948975

ABSTRACT

A series of pyrazole inhibitors of p38 mitogen-activated protein (MAP) kinase were designed using a binding model based on the crystal structure of 1 (SC-102) bound to p38 enzyme. New chemistry using dithietanes was developed to assemble nitrogen-linked substituents at the 5-position of pyrazoles. Calculated log D was used in tandem with structure-based design to guide medicinal chemistry strategy and improve the in vivo activity of a series of molecules. The crystal structure of an optimized inhibitor, 4 (SC-806), in complex with p38 enzyme was obtained to confirm the hypothesis that the addition of a basic nitrogen to the molecule induces an interaction with Asp112 of p38 alpha. A compound identified from this series was efficacious in an animal model of rheumatic disease.


Subject(s)
Antirheumatic Agents/chemical synthesis , Piperazines/chemical synthesis , Pyrazoles/chemical synthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Collagen , Crystallography, X-Ray , Male , Mice , Mice, Inbred DBA , Models, Molecular , Piperazines/chemistry , Piperazines/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats , Rats, Inbred Lew , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/chemistry
12.
Bioorg Med Chem Lett ; 15(5): 1315-9, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15713378

ABSTRACT

The synthesis and the preliminary expansion of this new class of CDK2 inhibitors are presented. The synthesis was accomplished using a solution-phase protocol amenable to rapid parallel expansion and suitable to be scaled-up in view of possible lead development. Following a medicinal chemistry program aimed at improving cell permeability and selectivity, a series of compounds with nanomolar activity in the biochemical assay and able to efficiently inhibit tumor cell proliferation has been obtained.


Subject(s)
CDC2-CDC28 Kinases/antagonists & inhibitors , Enzyme Inhibitors/classification , Enzyme Inhibitors/pharmacology , Pyrazoles/classification , Pyrazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallization , Crystallography, X-Ray , Cyclin A/antagonists & inhibitors , Cyclin-Dependent Kinase 2 , Enzyme Inhibitors/chemical synthesis , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Models, Molecular , Molecular Structure , Pyrazoles/chemical synthesis , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...