Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Am Chem Soc ; 145(44): 24035-24051, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37874670

ABSTRACT

Establishing a technological platform for creating clinical compounds inhibiting intracellular protein-protein interactions (PPIs) can open the door to many valuable drugs. Although small molecules and antibodies are mainstream modalities, they are not suitable for a target protein that lacks a deep cavity for a small molecule to bind or a protein found in intracellular space out of an antibody's reach. One possible approach to access these targets is to utilize so-called middle-size cyclic peptides (defined here as those with a molecular weight of 1000-2000 g/mol). In this study, we validated a new methodology to create oral drugs beyond the rule of 5 for intracellular tough targets by elucidating structural features and physicochemical properties for drug-like cyclic peptides and developing library technologies to afford highly N-alkylated cyclic peptide hits. We discovered a KRAS inhibitory clinical compound (LUNA18) as the first example of our platform technology.


Subject(s)
Peptides, Cyclic , Peptides, Cyclic/chemistry
2.
Int J Exp Pathol ; 98(4): 221-233, 2017 08.
Article in English | MEDLINE | ID: mdl-28895242

ABSTRACT

Hepatocellular carcinoma (HCC) is a common cancer worldwide and represents the outcome of the natural history of chronic liver disease. The growing rates of HCC may be partially attributable to increased numbers of people with non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). However, details of the liver-specific molecular mechanisms responsible for the NAFLD-NASH-HCC progression remain unclear, and mouse models that can be used to explore the exact factors that influence the progression of NAFLD/NASH to the more chronic stages of liver disease and subsequent HCC are not yet fully established. We have previously reported a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) as a dietary NASH model with rapidly progressive liver fibrosis in mice. The current study in C57BL/6J mice fed CDAHFD provided evidence for the chronic persistence of advanced hepatic fibrosis in NASH and disease progression towards HCC in a period of 36 weeks. When mice fed CDAHFD were switched back to a standard diet, hepatic steatosis was normalized and NAFLD activity score improved, but HCC incidence increased and the phenotype of fibrosis-associated HCC development was observed. Moreover, when mice continued to be fed CDAHFD for 60 weeks, HCC further developed without severe body weight loss or carcinogenesis in other organs. The autochthonous tumours showed a variety of histological features and architectural patterns including trabecular, pseudoglandular and solid growth. The CDAHFD mouse model might be a useful tool for studying the development of HCC from NAFLD/NASH, and potentially useful for better understanding pathological changes during hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular , Cell Transformation, Neoplastic , Choline Deficiency/metabolism , Diet, High-Fat , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/pathology , Animals , Choline/metabolism , Choline/pharmacology , Disease Models, Animal , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism
3.
Bioorg Med Chem Lett ; 19(13): 3426-9, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19481451

ABSTRACT

We successfully discovered peptidomimetic motilin antagonists (17c and 17d) through the improvement of physicochemical properties of a tetrapeptide antagonist (2). Furthermore, with oral administration and based on motilin antagonistic activity, both compounds suppressed motilin-induced colonic and gastric motility in conscious dogs.


Subject(s)
Gastrointestinal Agents/antagonists & inhibitors , Motilin/antagonists & inhibitors , Oligopeptides/chemical synthesis , Peptides/chemistry , Animals , Caco-2 Cells , Cell Line , Drug Discovery , Gastrointestinal Agents/metabolism , Humans , Motilin/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptides/chemical synthesis , Permeability , Rabbits , Rats
4.
Mol Cancer Ther ; 17(12): 2519-2529, 2018 12.
Article in English | MEDLINE | ID: mdl-30242093

ABSTRACT

Members of the tropomyosin receptor kinase (TRK) family are expressed in their constitutively activated forms as a result of a gene fusion that occurs across a wide variety of cancer types. We have identified CH7057288 as a potent and selective TRK inhibitor that belongs to a novel chemical class. CH7057288 showed selective inhibitory activity against TRKA, TRKB, and TRKC in cell-free kinase assays and suppressed proliferation of TRK fusion-positive cell lines, but not that of TRK-negative cell lines. Strong in vivo tumor growth inhibition was observed in subcutaneously implanted xenograft tumor models of TRK fusion-positive cells. Furthermore, in an intracranial implantation model mimicking brain metastasis, CH7057288 significantly induced tumor regression and improved event-free survival. Recently, resistant mutations in the kinase domain of TRK have been reported in patients who show disease progression after treatment with the TRK inhibitors now under clinical development. Our compound maintained similar levels of in vitro and in vivo activity against one of these resistant mutants as it did to wild-type TRK. An X-ray crystal structure of the TRKA and CH7057288 complex supported the activity against the mutant. In addition, gene expression analysis revealed that CH7057288 suppressed MAPK and E2F pathways as downstream signaling of TRK fusion. Therefore, CH7057288 could be a promising therapeutic agent for TRK fusion-positive cancer.


Subject(s)
Benzofurans/pharmacology , Neoplasms/pathology , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Mutation/genetics , Protein Kinases/chemistry , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
Int J Pharm ; 293(1-2): 183-92, 2005 Apr 11.
Article in English | MEDLINE | ID: mdl-15778056

ABSTRACT

The purpose of the present study was to examine the oral drug absorption predictability of the theoretical passive absorption model (TPAM). As chemical descriptors of drugs, the octanol/buffer distribution coefficient at pH 6.0 (D(ow)), intrinsic octanol-water partition coefficient (P(ow)), pK(a), and molecular weight (MW) were calculated from the chemical structure. Total passive intestinal membrane permeation consists of transcellular, paracellular and unstirred water layer (UWL) permeation. Transcellular permeation was modeled based on the pH-partition hypothesis with correction for cationic species permeation, and the independent variables were D(ow), P(ow), and pK(a). Paracellular permeation was modeled as a size-restricted diffusion within a negative electrostatic field-of-force, and the independent variables were MW and pK(a). UWL permeation was modeled as diffusion across a water layer, and the independent variable was MW. Cationic species permeation in the transcellular permeation model and the effect of a negative electric field-of-force in the paracellular permeation model were the extensions to the previous TPAM. The coefficients of the paracellular and UWL permeation models were taken from the literature. A data set of 258 compounds with observed values of Fa% (the fraction of a dose absorbed in humans) taken from the literature was employed to optimize four fitting coefficients in the transcellular permeation model. The TPAM predicted Fa%, with root mean square errors of 15-21% and a correlation coefficient (CC) of 0.78-0.88. In addition, the TPAM predicted the effective human intestinal membrane permeability with a CC of 0.67-0.77, as well as the contribution of paracellular permeation. The TPAM was found to predict oral absorption from the chemical structure of drugs with adequate predictability for usage in drug discovery.


Subject(s)
Intestinal Absorption/physiology , Models, Chemical , Pharmaceutical Preparations/metabolism , Administration, Oral , Humans , Intestinal Absorption/drug effects , Predictive Value of Tests
6.
Drug Metab Pharmacokinet ; 24(6): 500-10, 2009.
Article in English | MEDLINE | ID: mdl-20045985

ABSTRACT

A method of assessing the risk of drug-drug interaction (DDI) caused by mechanism-based inhibition (MBI) was developed for early-stage drug development using cytochrome P450 (CYP) 3A4 inhibition screening data. CYP3A4 inhibition was evaluated using a fluorescent substrate with or without preincubation containing an inhibitor. The results showed that five well-known mechanism-based inhibitors, but not the competitive inhibitor ketoconazole, had lower IC(50) after preincubation, suggesting the utility of the IC(50) shift by preincubation to discern mechanism-based inhibitors. A method to approximately predict the change in the area under the concentration-time curve (AUC) of a co-administered drug by MBI was found using IC(50) shift data and the unbound mean plasma concentration of the inhibitor. From our predictions of change in the AUC for 38 drugs using this method, all mechanism-based inhibitors causing change in the AUC of more than 200% were predicted to be high risk. In conclusion, our method provides a simple assessment of the risk of DDI from mechanism-based inhibitors, especially in the early stages of drug development.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Drug Interactions/physiology , Ketoconazole/pharmacology , Microsomes, Liver/drug effects , Algorithms , Area Under Curve , Computer Simulation , Cytochrome P-450 CYP3A , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Ketoconazole/pharmacokinetics , Kinetics , Models, Biological , Models, Statistical , Pharmaceutical Preparations/chemistry , Substrate Specificity
7.
Xenobiotica ; 37(12): 1421-32, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17926229

ABSTRACT

The objective was to investigate the mechanism of nonlinear pharmacokinetics of mitemcinal, the first acid-resistant non-peptide motilin agonist, in rats. Super-proportional increases of the cumulative rates of radioactivity excreted into bile and urine following oral administration of [3H]-mitemcinal suggested nonlinear absorption of mitemcinal in rats. To evaluate the fraction dose absorbed (Fa) and intestinal availability (Fg), [3H]-mitemcinal was orally administrated to rats, and tritium radioactivity and unchanged mitemcinal concentration were determined in the portal blood. Fa values for 0.2 mg/kg1, 0.5 mg/kg, and 5.0 mg/kg dose groups were 0.314, 0.353, and 0.569, respectively. Corresponding Fg values were 0.243, 0.296, and 0.513, respectively. In Caco-2 experiments, the permeation of [3H]-mitemcinal in the secretory direction was larger than in the absorptive direction and was inhibited by P-glycoprotein (P-gp) substrate digoxin. The results indicate that the saturation of P-gp-mediated membrane permeation and intestinal metabolism causes the nonlinear pharmacokinetics of mitemcinal in rats.


Subject(s)
Erythromycin/analogs & derivatives , Intestinal Mucosa/metabolism , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Administration, Oral , Animals , Bile/metabolism , Caco-2 Cells , Cell Membrane Permeability/drug effects , Digoxin/pharmacology , Dose-Response Relationship, Drug , Erythromycin/pharmacokinetics , Humans , Intestinal Absorption/drug effects , Male , Rats , Rats, Sprague-Dawley
8.
Pharm Res ; 23(6): 1144-56, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16715363

ABSTRACT

PURPOSE: The purpose of this study was to develop a new system for computer simulation to predict fraction absorbed (F(a)) of Biopharmaceutical Classification System (BCS) class II (low solubility-high permeability) drugs after oral administration to humans, from a miniscale dissolution test. METHODS: Human oral absorption of 12 lipophilic drugs was simulated theoretically by using the dissolution and permeation parameters of the drugs. A miniscale dissolution test and a solubility study were carried out in a conventional buffer and a biorelevant medium (pH 6.5). A dissolution parameter, which can simulate in vivo dissolution, was obtained from the in vitro dissolution curve. Human intestinal permeability was estimated assuming that the permeation was limited by diffusion through the unstirred water layer. The F(a) in humans was predicted and then compared with clinical data. RESULTS: The dissolution and solubility of most model drugs were faster and higher in a biorelevant medium than in a conventional buffer. The simulated absorption was limited by the drug dissolution rate and/or solubility. Predicted F(a) was significantly correlated with clinical data (correlation coefficient r2 = 0.82, p < 0.001) when the dissolution profiles in biorelevant medium were used for the simulation. CONCLUSIONS: This new system quantitatively simulated human absorption and would be beneficial for the prediction of human F(a) values for BCS class II drugs.


Subject(s)
Absorption , Biopharmaceutics , Computer Simulation , Models, Biological , Pharmaceutical Preparations/metabolism , Administration, Oral , Alkynes , Benzoxazines , Caco-2 Cells , Cyclopropanes , Griseofulvin/administration & dosage , Griseofulvin/chemistry , Griseofulvin/metabolism , Humans , Intestinal Absorption , Ivermectin/administration & dosage , Ivermectin/chemistry , Ivermectin/metabolism , Kinetics , Oxazines/administration & dosage , Oxazines/chemistry , Oxazines/metabolism , Permeability , Pharmaceutical Preparations/chemistry , Solubility , Water/chemistry
9.
Pharm Res ; 21(5): 749-55, 2004 May.
Article in English | MEDLINE | ID: mdl-15180329

ABSTRACT

PURPOSE: To improve predictions of fraction dose absorbed (Fa) for hydrophilic drugs, a correction of paracellular permeability using the pore radius of tight junctions (TJs) in Caco-2 monolayers was performed. METHODS: The apparent permeability coefficient (P9app)) of drugs was measured using the Caco-2 assay and the parallel artificial membrane permeation assay (PAMPA), and values were corrected with the pore radius of TJs. RESULTS: An equation for calculating the pore radius of TJs from the P(app) of lucifer yellow was obtained. The optimal pore radius of TJs in Caco-2 monolayers for predicting human Fa was calculated to be 7 A. The correlation between the actual and predicted Fa was improved by using the P(app) corrected with the pore radius of TJs. Permeability in the PAMPA, which was corrected using the pore radius and membrane potential, was well correlated with that in the Caco-2 assay. Most of the hydrophilic drugs tested in this study were absorbed mainly through the paracellular pathway. CONCLUSIONS: The results suggest the necessity of optimizing paracellular permeation for the prediction of Fa, and also the importance of the paracellular pathway to the absorption of hydrophilic drugs. This method might contribute to the setting of appropriate dosages and the development of hydrophilic drugs.


Subject(s)
Pharmaceutical Preparations/metabolism , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Absorption , Algorithms , Caco-2 Cells , Chemical Phenomena , Chemistry, Physical , Humans , Membranes, Artificial , Models, Biological , Permeability , Pharmaceutical Preparations/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL