Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 26(18): 4971-4983, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30145050

ABSTRACT

Janus kinases (JAKs) are considered promising targets for the treatment of autoimmune diseases including rheumatoid arthritis (RA) due to their important role in multiple cytokine receptor signaling pathways. Recently, several JAK inhibitors have been developed for the treatment of RA. Here, we describe the identification of the novel orally bioavailable JAK inhibitor 18, peficitinib (also known as ASP015K), which showed moderate selectivity for JAK3 over JAK1, JAK2, and TYK2 in enzyme assays. Chemical modification at the C4-position of lead compound 5 led to a large increase in JAK inhibitory activity and metabolic stability in liver microsomes. Furthermore, we determined the crystal structures of JAK1, JAK2, JAK3, and TYK2 in a complex with peficitinib, and revealed that the 1H-pyrrolo[2,3-b]pyridine-5-carboxamide scaffold of peficitinib forms triple hydrogen bonds with the hinge region. Interestingly, the binding modes of peficitinib in the ATP-binding pockets differed among JAK1, JAK2, JAK3, and TYK2. WaterMap analysis of the crystal structures suggests that unfavorable water molecules are the likely reason for the difference in orientation of the 1H-pyrrolo[2,3-b]pyridine-5-carboxamide scaffold to the hinge region among JAKs.


Subject(s)
Adamantane/analogs & derivatives , Drug Discovery , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/pharmacology , Niacinamide/analogs & derivatives , Adamantane/chemistry , Adamantane/pharmacokinetics , Adamantane/pharmacology , Adamantane/therapeutic use , Administration, Oral , Animals , Arthritis, Rheumatoid/drug therapy , Biological Availability , Humans , Janus Kinase Inhibitors/pharmacokinetics , Janus Kinase Inhibitors/therapeutic use , Mice , Niacinamide/chemistry , Niacinamide/pharmacokinetics , Niacinamide/pharmacology , Niacinamide/therapeutic use , Rats , Structure-Activity Relationship
2.
Bioorg Med Chem ; 26(12): 3716-3726, 2018 07 23.
Article in English | MEDLINE | ID: mdl-29887511

ABSTRACT

A new series of transient receptor potential vanilloid type 1 (TRPV1) antagonists were designed and synthesized from N-(3-hydroxyphenyl)-2-(piperidin-1-ylmethyl)biphenyl-4-carboxamide hydrochloride (8). SAR studies identified (R)-N-(1-methyl-2-oxo-1,2,3,4-tetrahydro-7-quinolyl)-2-[(2-methylpyrrolidin-1-yl)methyl]biphenyl-4-carboxamide hydrochloride (ASP8370, 7), as a compound with high aqueous solubility, satisfactory stability in human liver microsomes, and reduced CYP3A4 inhibition. ASP8370 was selected as a clinical development candidate with significant ameliorative effects on neuropathic pain. SAR studies also revealed the structural mechanisms underlying the switching between TRPV1 antagonism and agonism.


Subject(s)
Amides/chemistry , Drug Design , TRPV Cation Channels/antagonists & inhibitors , Administration, Oral , Amides/metabolism , Amides/therapeutic use , Anticonvulsants/chemical synthesis , Anticonvulsants/metabolism , Anticonvulsants/therapeutic use , Biphenyl Compounds/chemistry , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Microsomes, Liver/metabolism , Neuralgia/drug therapy , Solubility , Structure-Activity Relationship , TRPV Cation Channels/metabolism
3.
Bioorg Med Chem ; 25(20): 5311-5326, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28789911

ABSTRACT

Janus kinases (JAKs) play a crucial role in cytokine mediated signal transduction. JAK inhibitors have emerged as effective immunomodulative agents for the prevention of transplant rejection. We previously reported that the tricyclic imidazo-pyrrolopyridinone 2 is a potent JAK inhibitor; however, it had poor oral absorption due to low membrane permeability. Here, we report the structural modification of compound 2 into the tricyclic dipyrrolopyridine 18a focusing on reduction of polar surface area (PSA), which exhibits potent in vitro activity, improved membrane permeability and good oral bioavailability. Compound 18a showed efficacy in rat heterotopic cardiac transplants model.


Subject(s)
Adjuvants, Immunologic/pharmacology , Drug Discovery , Janus Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Administration, Oral , Animals , Biological Availability , Cell Membrane Permeability/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Graft Survival/drug effects , Heart Transplantation , Humans , Janus Kinases/metabolism , Male , Molecular Structure , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Pyridines/administration & dosage , Pyridines/chemistry , Pyrroles/administration & dosage , Pyrroles/chemistry , Rats , Rats, Inbred ACI , Rats, Inbred Lew , Rats, Sprague-Dawley , Structure-Activity Relationship
5.
J Pharmacol Sci ; 133(1): 25-33, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28117214

ABSTRACT

The Janus kinase (JAK) family of tyrosine kinases is associated with various cytokine receptors. JAK1 and JAK3 play particularly important roles in the immune response, and their inhibition is expected to provide targeted immune modulation. Several oral JAK inhibitors have recently been developed for treating autoimmune diseases, including rheumatoid arthritis (RA). Here, we investigated the pharmacological effects of peficitinib (formerly known as ASP015K), a novel, chemically synthesized JAK inhibitor. We found that peficitinib inhibited JAK1 and JAK3 with 50% inhibitory concentrations of 3.9 and 0.7 nM, respectively. Peficitinib also inhibited IL-2-dependent T cell proliferation in vitro and STAT5 phosphorylation in vitro and ex vivo. Furthermore, peficitinib dose-dependently suppressed bone destruction and paw swelling in an adjuvant-induced arthritis model in rats via prophylactic or therapeutic oral dosing regimens. Peficitinib also showed efficacy in the model by continuous intraperitoneal infusion. Area under the concentration versus time curve (AUC) at 50% inhibition of paw swelling via intraperitoneal infusion was similar to exposure levels of AUC at 50% inhibition via oral administration, implying that AUC might be important for determining the therapeutic efficacy of peficitinib. These data suggest that peficitinib has therapeutic potential for the oral treatment of RA.


Subject(s)
Adamantane/analogs & derivatives , Arthritis, Experimental/drug therapy , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Niacinamide/analogs & derivatives , Adamantane/administration & dosage , Adamantane/pharmacology , Adamantane/therapeutic use , Adjuvants, Immunologic/adverse effects , Administration, Oral , Animals , Arthritis, Experimental/chemically induced , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Infusions, Parenteral , Male , Niacinamide/administration & dosage , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phosphorylation/drug effects , Rats , STAT5 Transcription Factor/blood , STAT5 Transcription Factor/metabolism , T-Lymphocytes/physiology
6.
Eur J Pharmacol ; 796: 69-75, 2017 Feb 05.
Article in English | MEDLINE | ID: mdl-27993641

ABSTRACT

Janus family kinases (JAKs) are essential molecules for cytokine responses and attractive targets for the treatment of transplant rejection and autoimmune diseases. Several JAK inhibitors have shown demonstrable effects on acute rejection in experimental cardiac transplant models. However, little is known about the potential benefits of JAK inhibitors on chronic rejection outcomes such as vasculopathy and fibrosis. Here, we examined the pharmacological profile of a novel JAK inhibitor, AS2553627, and explored its therapeutic potential in chronic rejection as well as acute rejection in a rat cardiac transplant model. AS2553627 potently inhibited JAK kinases but showed no inhibition of other kinases, including TCR-associated molecules. The compound also suppressed proliferation of IL-2 stimulated human and rat T cells. In a rat cardiac transplant model, oral administration of AS2553627 alone or co-administration with a sub-therapeutic dose of tacrolimus effectively prolonged cardiac allograft survival, suggesting the efficacy in treating acute rejection. To evaluate the effect on chronic rejection, recipient rats were administered a therapeutic dose of tacrolimus for 90 days. In combination with tacrolimus, AS2553627 significantly reduced cardiac allograft vasculopathy and fibrosis that tacrolimus alone did not inhibit. AS2553627 at the effective dose in rat transplantation models did not significantly reduce reticulocyte counts in peripheral whole blood after in vivo erythropoietin administration, indicating a low risk for anemia. These results suggest that AS2553627 may be a therapeutic candidate for the prevention of not only acute but also chronic rejection in cardiac transplantation.


Subject(s)
Allografts , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Janus Kinases/antagonists & inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Allografts/pathology , Animals , Cell Count , Cell Proliferation/drug effects , Drug Interactions , Graft Rejection/blood , Graft Rejection/pathology , Humans , Reticulocytes/drug effects , Reticulocytes/pathology , Tacrolimus/pharmacology , Time Factors
8.
Bioorg Med Chem ; 24(19): 4711-4722, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27544589

ABSTRACT

In organ transplantation, T cell-mediated immune responses play a key role in the rejection of allografts. Janus kinase 3 (JAK3) is specifically expressed in hematopoietic cells and associated with regulation of T cell development via interleukin-2 signaling pathway. Here, we designed novel 4,6-diaminonicotinamide derivatives as immunomodulators targeting JAK3 for prevention of transplant rejection. Our optimization of C4- and C6-substituents and docking calculations to JAK3 protein confirmed that the 4,6-diaminonicotinamide scaffold resulted in potent inhibition of JAK3. We also investigated avoidance of human ether-a-go-go related gene (hERG) inhibitory activity. Selected compound 28 in combination with tacrolimus prevented allograft rejection in a rat heterotopic cardiac transplantation model.


Subject(s)
6-Aminonicotinamide/analogs & derivatives , 6-Aminonicotinamide/pharmacology , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , 6-Aminonicotinamide/chemical synthesis , 6-Aminonicotinamide/therapeutic use , Animals , Graft Rejection/prevention & control , Heart Transplantation , Humans , Immunologic Factors/chemical synthesis , Immunologic Factors/therapeutic use , Janus Kinase 3/metabolism , Molecular Docking Simulation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/therapeutic use , Rats
9.
Bioorg Med Chem ; 23(15): 4871-4883, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26071372

ABSTRACT

Janus kinases (JAKs) regulate various inflammatory and immune responses and are targets for the treatment of inflammatory and immune diseases. As a novel class of immunomodulators targeting JAK3, 1H-pyrrolo[2,3-b]pyridine-5-carboxamide derivatives are promising candidates for treating such diseases. In chemical modification of lead compound 2, the substitution of a cycloalkyl ring for an N-cyanopyridylpiperidine in C4-position was effective for increasing JAK3 inhibitory activity. In addition, modulation of physical properties such as molecular lipophilicity and basicity was important for reducing human ether-a-go-go-related gene (hERG) inhibitory activity. Our optimization study gave compound 31, which exhibited potent JAK3 inhibitory activity as well as weak hERG inhibitory activity. In cellular assay, 31 exhibited potent immunomodulating effect on IL-2-stimulated T cell proliferation. In a pharmacokinetic study, good metabolic stability and oral bioavailability of 31 were achieved in rats, dogs, and monkeys. Further, 31 prolonged graft survival in an in vivo rat heterotopic cardiac transplant model.


Subject(s)
Amides/chemistry , Immunologic Factors/chemical synthesis , Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Administration, Oral , Amides/pharmacokinetics , Amides/therapeutic use , Animals , Binding Sites , Cell Proliferation/drug effects , Dogs , Graft Rejection/prevention & control , Half-Life , Haplorhini , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Interleukin-2/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Janus Kinase 3/metabolism , Male , Microsomes, Liver/metabolism , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary , Pyridines/chemistry , Rats , Rats, Inbred Lew , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transplantation, Heterotopic
10.
Bioorg Med Chem ; 23(15): 4846-4859, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26059596

ABSTRACT

Because Janus kinases (JAKs) play a crucial role in cytokine-mediated signal transduction, JAKs are an attractive target for the treatment of organ transplant rejection and autoimmune diseases such as rheumatoid arthritis (RA). To identify JAK inhibitors, we focused on the 1H-pyrrolo[2,3-b]pyridine derivative 3, which exhibited moderate JAK3 and JAK1 inhibitory activities. Optimization of 3 identified the tricyclic imidazo-pyrrolopyridinone derivative 19, which exhibited potent JAK3 and JAK1 inhibitory activities (IC50=1.1 nM, 1.5 nM, respectively) with favorable metabolic stability.


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyridones/chemistry , Animals , Binding Sites , Cell Proliferation/drug effects , Cells, Cultured , Crystallography, X-Ray , Cytochrome P-450 CYP3A/metabolism , Humans , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Janus Kinase 3/metabolism , Male , Microsomes, Liver/metabolism , Molecular Docking Simulation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyridines/chemistry , Pyridones/chemical synthesis , Pyridones/pharmacology , Pyrroles/chemistry , Rats , Rats, Inbred Lew , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , Structure-Activity Relationship
11.
Chem Pharm Bull (Tokyo) ; 63(5): 341-53, 2015.
Article in English | MEDLINE | ID: mdl-25786493

ABSTRACT

Janus kinases (JAKs) have been known to play crucial roles in modulating a number of inflammatory and immune mediators. Here, we describe a series of 1H-pyrrolo[2,3-b]pyridine derivatives as novel immunomodulators targeting JAK3 for use in treating immune diseases such as organ transplantation. In the chemical modification of compound 6, the introduction of a carbamoyl group to the C5-position and substitution of a cyclohexylamino group at the C4-position of the 1H-pyrrolo[2,3-b]pyridine ring led to a large increase in JAK3 inhibitory activity. Compound 14c was identified as a potent, moderately selective JAK3 inhibitor, and the immunomodulating effect of 14c on interleukin-2-stimulated T cell proliferation was shown. Docking calculations and WaterMap analysis of the 1H-pyrrolo[2,3-b]pyridine-5-carboxamide derivatives were conducted to confirm the substituent effects on JAK3 inhibitory activity.


Subject(s)
Janus Kinase 3/antagonists & inhibitors , Pyridines/chemical synthesis , Pyridines/pharmacology , Animals , Cell Proliferation , Immunomodulation , Lymphocytes/drug effects , Lymphocytes/physiology , Male , Medical Informatics , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Protein Binding , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Spleen/cytology
12.
Tetrahedron ; 65(33): 6489-6509, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-20640040

ABSTRACT

Three syntheses of the architecturally complex, cytotoxic marine macrolide (+)-spongistatin 1 (1) are reported. Highlights of the first-generation synthesis include: use of a dithiane multicomponent linchpin coupling tactic for construction of the AB and CD spiroketals, and their union via a highly selective Evans boron-mediated aldol reaction en route to an ABCD aldehyde; introduction of the C(44)-C(51) side chain via a Lewis acid-mediated ring opening of a glucal epoxide with an allylstannane to assemble the EF subunit; and final fragment union via Wittig coupling of the ABCD and EF subunits to form the C(28)-C(29) olefin, followed by regioselective Yamaguchi macrolactonization and global deprotection. The second- and third- generation syntheses, designed with the goal of accessing one gram of (+)-spongistatin 1 (1), maintain both the first-generation strategy for the ABCD aldehyde and final fragment union, while incorporating two more efficient approaches for construction of the EF Wittig salt. The latter combine the original chelation-controlled dithiane union of the E- and F-ring progenitors with application of a highly efficient cyanohydrin alkylation to append the F-ring side chain, in conjunction with two independent tactics to access the F-ring pyran. The first F-ring synthesis showcases a Petasis-Ferrier union/rearrangement protocol to access tetrahydropyrans, permitting the preparation of 750 mgs of the EF Wittig salt, which in turn was converted to 80 mg of (+)-spongistatin 1, while the second F-ring strategy, incorporates an organocatalytic aldol reaction as the key construct, permitting completion of 1.009 g of totally synthetic (+)-spongistatin 1 (1). A brief analysis of the three syntheses alongside our earlier synthesis of (+)-spongistatin 2 is also presented.

13.
Org Lett ; 6(20): 3637-40, 2004 Sep 30.
Article in English | MEDLINE | ID: mdl-15387567

ABSTRACT

[structure: see text] An efficient, stereocontrolled, and scalable second-generation synthesis of (+)-3, an advanced EF subtarget for the total synthesis of (+)-spongistatin 1, has been achieved. Highlights of the strategy include preparation of the F-ring pyran via a Petasis-Ferrier union/rearrangement sequence and installation of the chlorodiene side chain employing a cyanohydrin alkylation. The longest linear sequence, 26 steps, proceeds in 8.3% overall yield.


Subject(s)
Macrolides/chemical synthesis , Animals , Catalysis , Cyclization , Indicators and Reagents , Macrolides/analysis , Molecular Structure , Porifera/chemistry , Stereoisomerism
14.
Org Lett ; 5(5): 761-4, 2003 Mar 06.
Article in English | MEDLINE | ID: mdl-12605509

ABSTRACT

A stereocontrolled, total synthesis of (+)-spongistatin 1 (1) has been achieved. Union of a second-generation EF Wittig salt (+)-3 with the advanced ABCD aldehyde (-)-4, followed by regioselective macrolactonization and global deprotection afforded (+)-spongistatin 1 (1). The longest linear sequence, 29 steps, proceeded in 0.5% overall yield.


Subject(s)
Antineoplastic Agents/chemical synthesis , Ethers, Cyclic/chemical synthesis , Lactones/chemical synthesis , Macrolides , Aldehydes/chemistry , Lactones/chemistry , Stereoisomerism
15.
J Org Chem ; 61(11): 3700-3705, 1996 May 31.
Article in English | MEDLINE | ID: mdl-11667218

ABSTRACT

Total synthesis of trideca-O-methyl-alpha-pedunculagin was achieved by a simple sequence. The key step is the synthesis of methyl 4,6-O-benzylidene-2,3-O-[(S)-4,4',5,5',6,6'-hexamethoxydiphenoyl]-alpha-D-glucopyranoside through intramolecular ester-cyclization of racemic hexamethoxydiphenoyl chloride with methyl 4,6-O-benzylidene-alpha-D-glucopyranoside at the 2,3-position. The diastereoselectivity results obtained in the intramolecular cyclization of hexamethoxydiphenic acid to the carbohydrate core raises a very interesting point in considering the pathway of (R)-diphenic acid biosynthesis.

SELECTION OF CITATIONS
SEARCH DETAIL
...