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










Database
Publication year range
1.
Nihon Yakurigaku Zasshi ; 158(3): 273-281, 2023.
Article in Japanese | MEDLINE | ID: mdl-37121712

ABSTRACT

On March 28th, 2022, asciminib hydrochloride (Scemblix® Tablets 20 |mg/40 |mg), the world's first tyrosine kinase inhibitor (TKI) specifically targeting the ABL myristoyl pocket (STAMP inhibitor), was approved for chronic myeloid leukemia (CML) resistant or intolerant to prior therapy. Asciminib specifically binds to the myristoyl pocket, an allosteric site of BCR::ABL1, and inhibits the ABL1 family molecules. In vitro and in vivo pharmacology studies demonstrated cell growth inhibition and antitumor effects of asciminib. The international phase I study for patients with chronic or accelerated phase CML investigated the maximum tolerated dose (MTD) and recommended dose for expansion (RDE) of asciminib monotherapy. However, the MTD was not reached, so and RDE was determined based on tolerability, safety, pharmacokinetics (PK) and preliminary efficacy data obtained by the time of the study. RDE was determined to be 40 |mg twice daily in chronic or accelerated phase CML without T315I mutation, and 200 |mg twice daily in chronic or accelerated phase CML with T315I mutation. The international phase III study for patients with chronic phase CML who were previously treated with ≥2 TKIs and resistant or intolerant to the recent treatment demonstrated the superiority of asciminib over bosutinib in achieving the primary endpoint of a major molecular response (MMR) at week 24. Regarding safety, the most common treatment-related adverse event in asciminib arm was thrombocytopenia, and others included neutropenia. Asciminib is expected to be a new treatment option for CML patients who have limited choices due to resistance or intolerance to previous therapies.


Subject(s)
Antineoplastic Agents , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use
2.
Am J Nephrol ; 48(3): 157-164, 2018.
Article in English | MEDLINE | ID: mdl-30176654

ABSTRACT

BACKGROUND: TP0463518 is a novel hypoxia-inducible factor prolyl hydroxylase inhibitor developed to aid in the treatment of anemia associated with chronic kidney disease (CKD) and is expected to increase erythropoietin (EPO) derived from liver. Two phase I studies were conducted in healthy volunteers (HV) and CKD patients undergoing hemodialysis (i.e., HD patients) or those not undergoing dialysis (i.e., ND patients). METHODS: Pharmacokinetics, pharmacodynamics, and safety profiles of TP0463518 were assessed. Forty HV received single oral doses of TP0463518 at 3, 6, 11, 20, and 36 mg or placebo. Twenty ND patients received single doses of TP0463518 at 1, 6, and 11 mg and 9 HD patients received TP0463518 at 1 and 11 mg doses. To identify the source organ of EPO, glycosylation patterns were determined using percentage migrated isoform (PMI) values. RESULTS: Declining renal function slowed elimination of TP0463518 and increased the mean AUC0-∞. ∆Emax of serum EPO in 11-mg groups of HV, ND patients, and HD patients were 24.37 ± 11.37, 201.57 ± 130.34, and 1,324.76 ± 1,189.24 mIU/mL respectively. A strong correlation was -observed between logarithm conversions of ∆Emax and AUC0-∞ with correlation coefficients of 0.945. PMI values of blood after TP0463518 administration were elevated to similar or higher levels in comparison with those of umbilical cord blood, which mainly contains liver-derived EPO. CONCLUSIONS: TP0463518 induced dose-dependent EPO production, mainly derived from the liver in HV and CKD patients. These results suggest that TP0463518 is a new strategy for treating anemia in CKD, which can be used regardless of renal functions.


Subject(s)
Anemia/drug therapy , Dihydropyridines/pharmacology , Erythropoietin/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Liver/drug effects , Pyridines/pharmacology , Renal Insufficiency, Chronic/complications , Administration, Oral , Adult , Aged , Anemia/blood , Area Under Curve , Dihydropyridines/therapeutic use , Dose-Response Relationship, Drug , Female , Glomerular Filtration Rate , Healthy Volunteers , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Kidney/metabolism , Kidney/physiopathology , Liver/metabolism , Male , Middle Aged , Pyridines/therapeutic use , Renal Dialysis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/therapy , Treatment Outcome , Young Adult
3.
Chem Pharm Bull (Tokyo) ; 64(11): 1630-1640, 2016.
Article in English | MEDLINE | ID: mdl-27803474

ABSTRACT

We previously identified 3-chloro-N-{(S)-[3-(1-ethyl-1H-pyrazol-4-yl)phenyl][(2S)-piperidine-2-yl]methyl}-4-(trifluoromethyl)pyridine-2-carboxamide (5, TP0439150) as a potent and orally available glycine transporter 1 (GlyT1) inhibitor. In this article, we describe our identification of 1-methyl-N-(propan-2-yl)-N-({2-[4-(trifluoromethoxy)phenyl]pyridin-4-yl}methyl)-1H-imidazole-4-carboxamide (7n) as a structurally diverse back-up compound of 5, using central nervous system multiparameter optimization (CNS MPO) as a drug-likeness guideline. Compound 7n showed a higher CNS MPO score and different physicochemical properties as compared to 5. Compound 7n exhibited potent GlyT1 inhibitory activity, a favorable pharmacokinetics profile, and elicited an increase in the cerebrospinal fluid (CSF) concentration of glycine in rats.


Subject(s)
Cerebrospinal Fluid/chemistry , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Imidazoles/pharmacology , Pyridines/pharmacology , Administration, Oral , Animals , Cerebrospinal Fluid/metabolism , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Molecular Structure , Pyridines/chemistry , Rats , Structure-Activity Relationship
4.
Chem Pharm Bull (Tokyo) ; 64(9): 1321-37, 2016.
Article in English | MEDLINE | ID: mdl-27581637

ABSTRACT

A novel glycine transporter 1 (GlyT1) inhibitor was designed by the superposition of different chemotypes to enhance its inhibitory activity. Starting from 2-chloro-N-{(S)-phenyl[(2S)-piperidin-2-yl]methyl}-3-(trifluoromethyl)benzamide (2, SSR504734), the introduction of heteroaromatic rings enabled an increase in the GlyT1 inhibitory activity. Subsequent optimization led to the identification of 3-chloro-N-{(S)-[3-(1-ethyl-1H-pyrazol-4-yl)phenyl][(2S)-piperidine-2-yl]methyl}-4-(trifluoromethyl)pyridine-2-carboxamide (7w), which showed a powerful GlyT1 inhibitory activity (IC50=1.8 nM), good plasma exposure and a plasma to brain penetration in rats that was sufficient to evaluate the compound's pharmacological properties. Compound 7w showed significant effects in several rodent models for schizophrenia without causing any undesirable central nervous system side effects.


Subject(s)
Drug Discovery , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship
5.
Psychopharmacology (Berl) ; 232(15): 2849-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25869273

ABSTRACT

RATIONALE: Since the hypofunction of the N-methyl-D-aspartate (NMDA) receptor is known to be involved in the pathophysiology of schizophrenia, the enhancement of NMDA receptor function through glycine modulatory sites is expected to be a useful approach for the treatment of schizophrenia. OBJECTIVES: We investigated the efficacy of a glycine transporter 1 (GlyT1) inhibitor that potentiates NMDA receptor function by increasing synaptic glycine levels in animal models for cognitive dysfunction and negative symptoms, both of which are poorly managed by current antipsychotics. RESULTS: A newly synthesized GlyT1 inhibitor, 3-chloro-N-{(S)-[3-(1-ethyl-1H-pyrazol-4-yl)phenyl][(2S)-piperidin-2-yl]methyl}-4-(trifluoromethyl)pyridine-2-carboxamide (TASP0315003) significantly improved cognitive deficit induced by MK-801 in the object recognition test in rats. Likewise, TASP0315003 significantly improved MK-801 impaired cognition in the social recognition test in rats and also enhanced social memory in treatment-naïve rats. In addition, repeated phencyclidine (PCP) treatment reduced the social interaction of paired mice, which may reflect negative symptoms such as social withdrawal, and both acute and sub-chronic treatment with TASP0315003 reversed the reduction in social interaction induced by PCP. Moreover, TASP0315003 additionally exhibited an antidepressant effect in the forced swimming test in rats. In contrast, TASP0315003 did not affect spontaneous locomotor activity or rotarod performance and did not induce catalepsy, indicating that TASP0315003 does not cause sedation or motor dysfunction, which is sometimes observed with the use of current antipsychotics. CONCLUSIONS: These results suggest that GlyT1 inhibitors including TASP0315003 may be useful for the treatment of cognitive dysfunction and the negative symptoms of schizophrenia without having undesirable central nervous system side effects.


Subject(s)
Cognition Disorders/drug therapy , Cognition/drug effects , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Memory/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/drug therapy , Social Behavior , Animals , Antidepressive Agents/therapeutic use , Antipsychotic Agents/pharmacology , Cognition Disorders/metabolism , Disease Models, Animal , Glycine/metabolism , Male , Mice , Phencyclidine , Rats , Schizophrenia/metabolism
6.
J Med Chem ; 53(8): 3247-61, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20302302

ABSTRACT

Derivatives of a novel scaffold, C-phenyl 1-thio-D-glucitol, were prepared and evaluated for sodium-dependent glucose cotransporter (SGLT) 2 and SGLT1 inhibition activities. Optimization of substituents on the aromatic rings afforded five compounds with potent and selective SGLT2 inhibition activities. The compounds were evaluated for in vitro human metabolic stability, human serum protein binding (SPB), and Caco-2 permeability. Of them, (1S)-1,5-anhydro-1-[5-(4-ethoxybenzyl)-2-methoxy-4-methylphenyl]-1-thio-D-glucitol (3p) exhibited potent SGLT2 inhibition activity (IC(50) = 2.26 nM), with 1650-fold selectivity over SGLT1. Compound 3p showed good metabolic stability toward cryo-preserved human hepatic clearance, lower SPB, and moderate Caco-2 permeability. Since 3p should have acceptable human pharmacokinetics (PK) properties, it could be a clinical candidate for treating type 2 diabetes. We observed that compound 3p exhibits a blood glucose lowering effect, excellent urinary glucose excretion properties, and promising PK profiles in animals. Phase II clinical trials of 3p (TS-071) are currently ongoing.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/chemical synthesis , Sodium-Glucose Transporter 2 Inhibitors , Sorbitol/analogs & derivatives , Sorbitol/chemical synthesis , Animals , Biological Availability , Blood Proteins/metabolism , CHO Cells , Caco-2 Cells , Cell Membrane Permeability , Cricetinae , Cricetulus , Dogs , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Microsomes, Liver/metabolism , Protein Binding , Rats , Rats, Zucker , Sodium-Glucose Transporter 2 , Sorbitol/pharmacology , Structure-Activity Relationship , Tissue Distribution
7.
Bioorg Med Chem ; 16(8): 4359-66, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18348906

ABSTRACT

In this paper, we describe the synthesis of (+)-(1R( *),2R( *))-2-[(1S( *))-1-amino-1-carboxy-2-(9H-xanthen-9-yl)-ethyl]-1-fluorocyclopropanecarboxylic acid (+)-16a, a compound, that is, fluorinated at the alpha position of the carboxylic acid in the cyclopropane ring of a group II mGluRs antagonist, 1 (LY341495), using a previously reported stereoselective cyclopropanation reaction. The fluorinated compound (+)-16a exhibited almost the same affinity (IC(50)=3.49 nM) for mGluR2 as 1 but had a superior pharmacokinetic profile. Furthermore, a marked elevation of the plasma levels of (+)-16a was observed following the administration of a prodrug, (+)-17.


Subject(s)
Receptors, Metabotropic Glutamate/antagonists & inhibitors , Xanthenes/chemical synthesis , Xanthenes/pharmacology , Animals , Crystallography, X-Ray , Liver/drug effects , Liver/metabolism , Male , Models, Molecular , Molecular Structure , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacology , Protein Binding , Rats , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship , Xanthenes/chemistry
8.
Drug Metab Dispos ; 34(3): 369-74, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16326817

ABSTRACT

MGS0039 (3-(3,4-dichlorobenzyloxy)-2-amino-6-fluorobicyclo-[3.1.0]hexane-2,6-dicarboxylic acid) has been identified as a potent and selective antagonist for metabotropic glutamate receptors. However, the oral bioavailability of MGS0039 is 10.9% in rats, due to low absorption. Several prodrugs, synthesized to improve absorption, exhibited 40 to 70% bioavailability in rats. This study investigated in vitro metabolism using liver S9 fractions from both cynomolgus monkeys and humans and oral bioavailability in cynomolgus monkeys to select the prodrug most likely to exhibit optimal pharmacokinetic profiles in humans. In monkeys, transformation to active substance was observed (5.9-72.8%) in liver S9 fractions, and n-butyl, n-pentyl, 3-methylbutyl, and 4-methylpentyl ester prodrugs exhibited high transformation ratios (>64%). Cmax levels and F values after oral dosing increased to 4.1- to 6.3-fold and 2.4- to 6.3-fold, respectively, and a close relationship between transformation ratios and Cmax and F values was observed, indicating that the hydrolysis rate in liver S9 fractions is the key factor in determining oral bioavailability in monkeys. In humans, n-hexyl, n-heptyl, n-octyl, 5-methylbutyl, and 6-methylpentyl ester prodrugs exhibited high transformation ratios (>65%) in liver S9 fractions. With these prodrugs, n-hexyl, n-heptyl, and 5-methylpentyl ester, almost complete recovery (96-99%) was obtained. Given the transformation ratio, we anticipated that the n-heptyl alkyl ester prodrug would exhibit the highest oral bioavailability of active substances in humans, if the hydrolysis rate in liver S9 fractions is indeed the key factor in determining oral bioavailability in humans. On this basis, MGS0210 (3-(3,4-dichlorobenzyloxy)-2-amino-6-fluorobicyclo[3.1.0]hexane-2,6-dicarboxylic acid n-heptyl ester) seems to be a promising candidate among MGS0039 prodrugs.


Subject(s)
Bridged Bicyclo Compounds/pharmacokinetics , Dicarboxylic Acids/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Liver/metabolism , Prodrugs/pharmacokinetics , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Biological Availability , Bridged Bicyclo Compounds/blood , Bridged Bicyclo Compounds/metabolism , Dicarboxylic Acids/blood , Dicarboxylic Acids/metabolism , Excitatory Amino Acid Antagonists/blood , Excitatory Amino Acid Antagonists/metabolism , Humans , In Vitro Techniques , Macaca fascicularis , Male , Prodrugs/metabolism
9.
Biol Pharm Bull ; 26(1): 19-23, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12520165

ABSTRACT

The mechanism of the elevation of intracellular glutathione induced by low-dose gamma-rays was examined in RAW 264.7 cells. The expression of mRNA for gamma-glutamylcysteine synthetase (gamma-GCS) increased soon after gamma-ray (0.5 Gy) irradiation, and peaked between 3 h and 6 h post-irradiation. A dose of 0.25 to 0.5 Gy was optimum for induction of gamma-GCS mRNA expression at 3 h post-irradiation. The effect of inhibitors of activator protein-1 (AP-1) and nuclear factor kappaB (NF-kappaB) on the radiation-induced gamma-GCS gene expression was then examined. The induction of gamma-GCS mRNA expression was significantly suppressed when AP-1 DNA binding, but not NF-kappaB DNA binding, was inhibited. Finally, electrophoretic mobility shift assay showed that the low-dose radiation markedly increased the DNA binding of AP-1, but not NF-kappaB, soon after irradiation. These results suggest that the increase of glutathione levels in RAW 264.7 cells by low-dose gamma-ray irradiation is mediated by transcriptional regulation of the gamma-GCS gene, predominantly through the AP-1 binding site in its promoter.


Subject(s)
Gamma Rays , Glutathione/biosynthesis , Glutathione/radiation effects , Intracellular Fluid/radiation effects , Transcription Factor AP-1/physiology , Transcription Factor AP-1/radiation effects , Animals , Cell Line , Dose-Response Relationship, Radiation , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , Intracellular Fluid/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...