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
2.
Sci Rep ; 11(1): 11066, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34040108

ABSTRACT

The nuclear receptor retinoid-related orphan receptor gamma t (RORγt) plays a critical role in driving Th17 cell differentiation and expansion, as well as IL-17 production in innate and adaptive immune cells. The IL-23/IL-17 axis is implicated in several autoimmune and inflammatory diseases, and biologics targeting IL-23 and IL-17 have shown significant clinical efficacy in treating psoriasis and psoriatic arthritis. JNJ-61803534 is a potent RORγt inverse agonist, selectively inhibiting RORγt-driven transcription versus closely-related family members, RORα and RORß. JNJ-61803534 inhibited IL-17A production in human CD4+ T cells under Th17 differentiation conditions, but did not inhibit IFNγ production under Th1 differentiation conditions, and had no impact on in vitro differentiation of regulatory T cells (Treg), nor on the suppressive activity of natural Tregs. In the mouse collagen-induced arthritis model, JNJ-61803534 dose-dependently attenuated inflammation, achieving ~ 90% maximum inhibition of clinical score. JNJ-61803534 significantly inhibited disease score in the imiquimod-induced mouse skin inflammation model, and dose-dependently inhibited the expression of RORγt-regulated genes, including IL-17A, IL-17F, IL-22 and IL-23R. Preclinical 1-month toxicity studies in rats and dogs identified doses that were well tolerated supporting progression into first-in-human studies. An oral formulation of JNJ-61803534 was studied in a phase 1 randomized double-blind study in healthy human volunteers to assess safety, pharmacokinetics, and pharmacodynamics. The compound was well tolerated in single ascending doses (SAD) up to 200 mg, and exhibited dose-dependent increases in exposure upon oral dosing, with a plasma half-life of 164 to 170 h. In addition, dose-dependent inhibition of ex vivo stimulated IL-17A production in whole blood was observed, demonstrating in vivo target engagement. In conclusion, JNJ-61803534 is a potent and selective RORγt inhibitor that exhibited acceptable preclinical safety and efficacy, as well as an acceptable safety profile in a healthy volunteer SAD study, with clear evidence of a pharmacodynamic effect in humans.


Subject(s)
Anti-Inflammatory Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Gene Expression Regulation/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Inflammation/drug therapy , Inflammation/metabolism , Interleukins/genetics , Interleukins/metabolism , Mice , Skin Diseases/drug therapy , Skin Diseases/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism
3.
J Med Chem ; 63(17): 9181-9196, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32787105

ABSTRACT

Selective inhibitors of the GluN2B subunit of N-methyl-d-aspartate receptors in the ionotropic glutamate receptor superfamily have been targeted for the treatment of mood disorders. We sought to identify structurally novel, brain penetrant, GluN2B-selective inhibitors suitable for evaluation in a clinical setting in patients with major depressive disorder. We identified a new class of negative allosteric modulators of GluN2B that contain a 1,3-dihydro-imidazo[4,5-b]pyridin-2-one core. This series of compounds had poor solubility properties and poor permeability, which was addressed utilizing two approaches. First, a series of structural modifications was conducted which included replacing hydrogen bond donor groups. Second, enabling formulation development was undertaken in which a stable nanosuspension was identified for lead compound 12. Compound 12 was found to have robust target engagement in rat with an ED70 of 1.4 mg/kg. The nanosuspension enabled sufficient margins in preclinical toleration studies to nominate 12 for progression into advanced good laboratory practice studies.


Subject(s)
Antipsychotic Agents/chemical synthesis , Drug Design , Imidazoles/chemistry , Pyridines/chemistry , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Allosteric Regulation , Animals , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/therapeutic use , Brain/metabolism , Dogs , Drug Evaluation, Preclinical , Half-Life , Humans , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Male , Mood Disorders/drug therapy , Mood Disorders/pathology , Nanostructures/chemistry , Permeability/drug effects , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Solubility , Structure-Activity Relationship
4.
J Med Chem ; 61(1): 207-223, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29211470

ABSTRACT

A single pot dipolar cycloaddition reaction/Cope elimination sequence was developed to access novel 1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridine P2X7 antagonists that contain a synthetically challenging chiral center. The structure-activity relationships of the new compounds are described. Two of these compounds, (S)-(2-fluoro-3-(trifluoromethyl)phenyl)(1-(5-fluoropyrimidin-2-yl)-6-methyl-1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridin-5-yl)methanone (compound 29) and (S)-(3-fluoro-2-(trifluoromethyl)pyridin-4-yl)(1-(5-fluoropyrimidin-2-yl)-6-methyl-1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridin-5-yl)methanone (compound 35), were found to have robust P2X7 receptor occupancy at low doses in rat with ED50 values of 0.06 and 0.07 mg/kg, respectively. Compound 35 had notable solubility compared to 29 and showed good tolerability in preclinical species. Compound 35 was chosen as a clinical candidate for advancement into phase I clinical trials to assess safety and tolerability in healthy human subjects prior to the initiation of proof of concept studies for the treatment of mood disorders.


Subject(s)
Drug Design , Purinergic P2X Receptor Antagonists/chemical synthesis , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Receptors, Purinergic P2X7/metabolism , Animals , Cycloaddition Reaction , Dogs , Humans , Male , Mice , Models, Molecular , Molecular Conformation , Purinergic P2X Receptor Antagonists/chemistry , Purinergic P2X Receptor Antagonists/pharmacokinetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Rats , Stereoisomerism , Tissue Distribution
5.
J Med Chem ; 60(11): 4559-4572, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28493698

ABSTRACT

The synthesis and preclinical characterization of novel 4-(R)-methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridines that are potent and selective brain penetrant P2X7 antagonists are described. Optimization efforts based on previously disclosed unsubstituted 6,7-dihydro-4H-triazolo[4,5-c]pyridines, methyl substituted 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyrazines, and several other series lead to the identification of a series of 4-(R)-methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridines that are selective P2X7 antagonists with potency at the rodent and human P2X7 ion channels. These novel P2X7 antagonists have suitable physicochemical properties, and several analogs have an excellent pharmacokinetic profile, good partitioning into the CNS and show robust in vivo target engagement after oral dosing. Improvements in metabolic stability led to the identification of JNJ-54175446 (14) as a candidate for clinical development. The drug discovery efforts and strategies that resulted in the identification of the clinical candidate are described herein.


Subject(s)
Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/pharmacology , Receptors, Purinergic P2X7/drug effects , Animals , Biological Availability , Humans , Purinergic P2X Receptor Antagonists/pharmacokinetics
6.
J Med Chem ; 59(18): 8535-48, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27548392

ABSTRACT

The synthesis and SAR of a series of 4,5,6,7-tetrahydro-imidazo[4,5-c]pyridine P2X7 antagonists are described. Addressing P2X7 affinity and liver microsomal stability issues encountered with this template afforded methyl substituted 4,5,6,7-tetrahydro-imidazo[4,5-c]pyridines ultimately leading to the identification of 1 (JNJ 54166060). 1 is a potent P2X7 antagonist with an ED50 = 2.3 mg/kg in rats, high oral bioavailability and low-moderate clearance in preclinical species, acceptable safety margins in rats, and a predicted human dose of 120 mg of QD. Additionally, 1 possesses a unique CYP profile and was found to be a regioselective inhibitor of midazolam CYP3A metabolism.


Subject(s)
Purinergic P2X Receptor Antagonists/chemistry , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Receptors, Purinergic P2X7/metabolism , Administration, Oral , Animals , Dogs , Halogenation , Haplorhini , Humans , Imidazoles/administration & dosage , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Mice , Models, Molecular , Purinergic P2X Receptor Antagonists/administration & dosage , Purinergic P2X Receptor Antagonists/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Rats
7.
J Pharm Sci ; 105(2): 460-475, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26869412

ABSTRACT

Modification of biopharmaceutical molecules by covalent conjugation of polyethylene glycol (PEG) molecules is known to enhance pharmacologic and pharmaceutical properties of proteins and other large molecules and has been used successfully in 12 approved drugs. Both linear and branched-chain PEG reagents with molecular sizes of up to 40 kDa have been used with a variety of different PEG derivatives with different linker chemistries. This review describes the properties of PEG itself, the history and evolution of PEGylation chemistry, and provides examples of PEGylated drugs with an established medical history. A trend toward the use of complex PEG architectures and larger PEG polymers, but with very pure and well-characterized PEG reagents is described. Nonclinical toxicology findings related to PEG in approved PEGylated biopharmaceuticals are summarized. The effect attributed to the PEG part of the molecules as observed in 5 of the 12 marketed products was cellular vacuolation seen microscopically mainly in phagocytic cells which is likely related to their biological function to absorb and remove particles and macromolecules from blood and tissues. Experience with marketed PEGylated products indicates that adverse effects in toxicology studies are usually related to the active part of the drug but not to the PEG moiety.


Subject(s)
Biopharmaceutics/methods , Drug Approval , Pharmaceutical Preparations/chemistry , Polyethylene Glycols/chemistry , Animals , Chemistry, Pharmaceutical , Drug-Related Side Effects and Adverse Reactions/metabolism , Humans , Pharmaceutical Preparations/metabolism , Polyethylene Glycols/adverse effects , Polyethylene Glycols/metabolism , Protein Stability
8.
Br J Pharmacol ; 172(16): 4078-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25988595

ABSTRACT

BACKGROUND AND PURPOSE: Small-molecule inhibitors of prolyl hydroxylase (PHD) enzymes are a novel target for the treatment of anaemia and functional iron deficiency (FID). Other than being orally bioavailable, the differentiation of PHD inhibitors from recombinant human erythropoietin (rhEPO) has not been demonstrated. EXPERIMENTAL APPROACH: JNJ-42905343 was identified and characterized as a novel inhibitor of PHD and its action was compared with rhEPO in healthy rats and in a rat model of inflammation-induced anaemia and FID [peptidoglycan-polysaccharide (PGPS) model]. KEY RESULTS: Oral administration of JNJ-42905343 to healthy rats increased the gene expression of cytochrome b (DcytB) and divalent metal-ion transporter 1 (DMT1) in the duodenum, and increased plasma EPO. Repeated administration of JNJ-42905343 for 28 days increased blood haemoglobin, mean corpuscular haemoglobin (MCH) and mean corpuscular volume (MCV). The serum iron concentration was increased with low doses (0.3 mg·kg(-1) ) but reduced at high doses (6 mg·kg(-1) ). In PGPS-treated rats, administration of JNJ-42905343 for 28 days corrected FID and anaemia, as reflected by increased blood haemoglobin, MCH and MCV. Increased expression of DcytB and DMT1 genes in the duodenum resulting in increased iron availability was defined as the mechanism for these effects. rhEPO did not affect DcytB and DMT1 and was not effective in PGPS-treated rats. CONCLUSIONS AND IMPLICATIONS: PHD inhibition has a beneficial effect on iron metabolism in addition to stimulating the release of EPO. Small-molecule inhibitors of PHD such as JNJ-42905343 represent a mechanism distinct from rhEPO to treat anaemia and FID.


Subject(s)
Anemia/drug therapy , Prolyl-Hydroxylase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Quinazolinones/therapeutic use , Anemia/blood , Anemia/genetics , Anemia/metabolism , Animals , Cation Transport Proteins/genetics , Cytochromes b/genetics , Duodenum/drug effects , Duodenum/metabolism , Erythrocyte Indices/drug effects , Erythropoietin/blood , Erythropoietin/genetics , Erythropoietin/pharmacology , Erythropoietin/therapeutic use , Female , Gene Expression/drug effects , Hemoglobins/analysis , Inflammation/blood , Inflammation/chemically induced , Inflammation/complications , Inflammation/genetics , Inflammation/metabolism , Iron/blood , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Peptidoglycan , Prolyl-Hydroxylase Inhibitors/pharmacokinetics , Prolyl-Hydroxylase Inhibitors/pharmacology , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Quinazolinones/pharmacokinetics , Quinazolinones/pharmacology , Rats, Inbred Lew
SELECTION OF CITATIONS
SEARCH DETAIL
...