Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
J Clin Lipidol ; 17(3): 392-400, 2023.
Article in English | MEDLINE | ID: mdl-37164838

ABSTRACT

BACKGROUND: Inclisiran is a novel siRNA therapy that inhibits the synthesis of proprotein convertase subtilisin-kexin type 9 (PCSK9) by targeting the PCSK9 mRNA, consequently, decreases low-density lipoprotein cholesterol (LDL-C). OBJECTIVE: To assess the safety, PK and LDL-C lowering effects of inclisiran in the Chinese patients with elevated LDL-C despite treatment with maximally tolerated LDL-C lowering therapies. METHODS: Forty Chinese patients with hypercholesterolemia (LDL-C ≥100 mg/dL) who were on maximally tolerated statin were randomized to receive a single dose of either inclisiran sodium 100 or 300mg s.c. injection (each for 15 patients) or placebo (10 patients). Safety, pharmacokinetics and pharmacodynamics (i.e., PCSK9 and LDL-C levels) were evaluated for up to 90 days after the s.c. injection of study drug. RESULTS: Following single subcutaneous injections inclisiran sodium at 100 mg or 300 mg, inclisiran has a relative short elimination half-life (T1/2, 6.5 hours). Both plasma PCSK9 and serum LDL-C decreased rapidly and consistently, with the maximal reduction between Day 30 and Day 60; then the decreases of PCSK9 and LDL-C were generally maintained up to 56.4% and 49.6% of 100 mg, 74.9% and 58.3% of 300 mg, respectively, at day 90. All adverse events were mild or moderate in severity, and no discontinuations due to adverse events. There were no serious adverse events being reported. CONCLUSIONS: Inclisiran was generally safe and well tolerated. Single dose of both Inclisiran 100 and 300 mg significantly reduced PCSK9 and LDL-C levels in Chinese patients up to Day 90. The greatest reductions were observed with the 300 mg regimen of Inclisiran. TRIAL REGISTRATION: ClinicalTrials.gov: NCT04774003.


Subject(s)
Anticholesteremic Agents , Hypercholesterolemia , RNA, Small Interfering , Humans , Anticholesteremic Agents/therapeutic use , Cholesterol, LDL , East Asian People , Hypercholesterolemia/drug therapy , Proprotein Convertase 9/genetics , RNA, Small Interfering/therapeutic use
2.
J Med Chem ; 65(13): 8713-8734, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35790118

ABSTRACT

The "free drug hypothesis" assumes that, in the absence of transporters, the steady state free plasma concentrations equal to that at the site of action that elicit pharmacologic effects. While it is important to utilize the free drug hypothesis, exceptions exist that the free plasma exposures, either at Cmax, Ctrough, and Caverage, or at other time points, cannot represent the corresponding free tissue concentrations. This "drug concentration asymmetry" in both total and free form can influence drug disposition and pharmacological effects. In this review, we first discuss options to assess total and free drug concentrations in tissues. Then various drug design strategies to achieve concentration asymmetry are presented. Last, the utilities of tissue concentrations in understanding exposure-effect relationships and translational projections to humans are discussed for several therapeutic areas and modalities. A thorough understanding in plasma and tissue exposures correlation with pharmacologic effects can provide insightful guidance to aid drug discovery.


Subject(s)
Drug Discovery , Plasma , Humans , Membrane Transport Proteins
3.
J Pers ; 90(4): 631-644, 2022 08.
Article in English | MEDLINE | ID: mdl-34714936

ABSTRACT

OBJECTIVE: Laughter conveys important information that supports social communication and bonding. Research suggests that unique acoustic properties distinguish laughter that promotes affiliation from laughter that conveys dominance, but little is known about potential individual differences in laughter interpretation or contagion based on these specified social functions of laughter. Psychopathy is associated with both affiliative deficits (e.g., lack of empathy and impaired social bonding) and behaviors that assert social dominance (e.g., manipulativeness). Thus, relationships between psychopathic traits and impaired laughter interpretation or contagion could give insight into etiological pathways to psychopathy. METHOD: In two studies conducted with four independent samples (total N = 770), participants categorized laughter clips that varied in the degree of affiliation or dominance conveyed. RESULTS: Participants overall drew rich and accurate social inferences from dominant and affiliative laughter and modulated their interest in joining in with laughter based on the type and degree of affiliation and dominance conveyed. However, individuals higher in psychopathic traits failed to distinguish between laughter types and did not modulate their level of engagement based on laughter features. CONCLUSIONS: The results suggest a potential mechanism that underlies the broader social difficulties associated with psychopathic traits.


Subject(s)
Laughter , Antisocial Personality Disorder , Cues , Empathy , Humans , Social Dominance
4.
Drug Metab Dispos ; 48(10): 873-885, 2020 10.
Article in English | MEDLINE | ID: mdl-32665418

ABSTRACT

Capmatinib (INC280), a highly selective and potent inhibitor of the MET receptor tyrosine kinase, has demonstrated clinically meaningful efficacy and a manageable safety profile in patients with advanced non-small-cell lung cancer harboring MET exon 14-skipping mutations. We investigated the absorption, distribution, metabolism, and excretion of capmatinib in six healthy male volunteers after a single peroral dose of 600 mg 14C-labeled capmatinib. The mass balance, blood and plasma radioactivity, and plasma capmatinib concentrations were determined along with metabolite profiles in plasma, urine, and feces. The metabolite structures were elucidated using mass spectrometry and comparing with reference compounds. The parent compound accounted for most of the radioactivity in plasma (42.9% ± 2.9%). The extent of oral absorption was estimated to be 49.6%; the Cmax of capmatinib in plasma was reached at 2 hours (median time to reach Cmax). The apparent mean elimination half-life of capmatinib in plasma was 7.84 hours. Apparent distribution volume of capmatinib during the terminal phase was moderate-to-high (geometric mean 473 l). Metabolic reactions involved lactam formation, hydroxylation, N-dealkylation, formation of a carboxylic acid, hydrogenation, N-oxygenation, glucuronidation, and combinations thereof. M16, the most abundant metabolite in plasma, urine, and feces was formed by lactam formation. Absorbed capmatinib was eliminated mainly by metabolism and subsequent biliary/fecal and renal excretion. Excretion of radioactivity was complete after 7 days. CYP phenotyping demonstrated that CYP3A was the major cytochrome P450 enzyme subfamily involved in hepatic microsomal metabolism, and in vitro studies in hepatic cytosol indicated that M16 formation was mainly catalyzed by aldehyde oxidase. SIGNIFICANCE STATEMENT: The absorption, distribution, metabolism, and excretion of capmatinib revealed that capmatinib had substantial systemic availability after oral administration. It was also extensively metabolized and largely distributed to the peripheral tissue. Mean elimination half-life was 7.84 hours. The most abundant metabolite, M16, was formed by imidazo-triazinone formation catalyzed by cytosolic aldehyde oxidase. Correlation analysis, specific inhibition, and recombinant enzymes phenotyping demonstrated that CYP3A is the major enzyme subfamily involved in the hepatic microsomal metabolism of [14C]capmatinib.


Subject(s)
Aldehyde Oxidase/metabolism , Benzamides/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Imidazoles/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Triazines/pharmacokinetics , Administration, Oral , Benzamides/administration & dosage , Benzamides/adverse effects , Biotransformation , Cytosol/metabolism , Healthy Volunteers , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Intestinal Absorption , Male , Microsomes, Liver , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Tissue Distribution , Triazines/administration & dosage , Triazines/adverse effects
5.
Invest New Drugs ; 38(6): 1774-1783, 2020 12.
Article in English | MEDLINE | ID: mdl-32410080

ABSTRACT

Background Overcoming resistance to anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (mAbs) in patients with KRAS wildtype (WT) metastatic colorectal cancer (mCRC) could help meet the needs of patients with limited treatment options. Methods In this phase 1b study, patients with N/KRAS WT, MET-positive mCRC who had progressed following anti-EGFR mAb treatment received escalating oral doses of capmatinib (150, 300, and 400 mg) twice daily plus weekly intravenous cetuximab (at the approved dose). The primary objective was to establish a recommended dose for expansion (RDE) of capmatinib in combination with cetuximab. Safety, preliminary activity, pharmacokinetics, and pharmacodynamics were also explored. Results Thirteen patients were enrolled. No patients experienced a dose-limiting toxicity at investigated doses; the RDE was established as capmatinib 400 mg twice daily plus cetuximab. All patients experienced adverse events (AEs) suspected to be related to the study treatment. Five patients (38.5%) reported study-drug-related AEs of grade 3/4 in severity. No patients achieved a complete or partial response according to RECIST v1.1; however, tumor shrinkage of 29-44% was observed in 4 patients. Conclusions Capmatinib plus cetuximab was well tolerated. Preliminary signs of activity were observed. Further investigation is warranted to obtain efficacy data and refine predictive biomarkers of response. Clinical trial registration NCT02205398.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzamides/administration & dosage , Cetuximab/administration & dosage , Colorectal Neoplasms/drug therapy , Imidazoles/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Triazines/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Benzamides/adverse effects , Benzamides/pharmacokinetics , Cell Line, Tumor , Cetuximab/adverse effects , Cetuximab/pharmacokinetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Female , Head and Neck Neoplasms/drug therapy , Humans , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met/metabolism , Response Evaluation Criteria in Solid Tumors , Squamous Cell Carcinoma of Head and Neck/drug therapy , Triazines/adverse effects , Triazines/pharmacokinetics , ras Proteins/genetics
6.
Drug Discov Today ; 25(3): 491-496, 2020 03.
Article in English | MEDLINE | ID: mdl-31926136

ABSTRACT

Some Asian regulators currently require Phase I data in Asians before joining global Phase II/III trials. Here, we discuss inherent limitations of Phase I ethnic sensitivity studies (ESS) to identify potential interethnic differences. We review recent new drug applications (NDAs) for Japan and China to critically assess the value of separate ESSs in Asian populations. Given that the observed value of ESS was limited, we propose a new global drug development paradigm: if relevant safety, pharmacokinetic (PK), and pharmacogenetic (PG) data are available from the original Phase I study population, it might be possible to extrapolate those data to Asian populations for their inclusion in Phase II/III trials, without an ESS. This could help to streamline drug development in Asia while still addressing regulatory requirements.


Subject(s)
Asian People , Clinical Trials, Phase I as Topic/methods , Drug Development/methods , China , Clinical Trials, Phase I as Topic/legislation & jurisprudence , Clinical Trials, Phase II as Topic/legislation & jurisprudence , Clinical Trials, Phase III as Topic/legislation & jurisprudence , Drug Development/legislation & jurisprudence , Ethnicity , Humans , Japan
7.
J Neurooncol ; 146(1): 79-89, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31776899

ABSTRACT

PURPOSE: To estimate the maximum tolerated dose (MTD) and/or identify the recommended Phase II dose (RP2D) for combined INC280 and buparlisib in patients with recurrent glioblastoma with homozygous phosphatase and tensin homolog (PTEN) deletion, mutation or protein loss. METHODS: This multicenter, open-label, Phase Ib/II study included adult patients with glioblastoma with mesenchymal-epithelial transcription factor (c-Met) amplification. In Phase Ib, patients received INC280 as capsules or tablets in combination with buparlisib. In Phase II, patients received INC280 only. Response was assessed centrally using Response Assessment in Neuro-Oncology response criteria for high-grade gliomas. All adverse events (AEs) were recorded and graded. RESULTS: 33 patients entered Phase Ib, 32 with altered PTEN. RP2D was not declared due to potential drug-drug interactions, which may have resulted in lack of efficacy; thus, Phase II, including 10 patients, was continued with INC280 monotherapy only. Best response was stable disease in 30% of patients. In the selected patient population, enrollment was halted due to limited activity with INC280 monotherapy. In Phase Ib, the most common treatment-related AEs were fatigue (36.4%), nausea (30.3%) and increased alanine aminotransferase (30.3%). MTD was identified at INC280 Tab 300 mg twice daily + buparlisib 80 mg once daily. In Phase II, the most common AEs were headache (40.0%), constipation (30.0%), fatigue (30.0%) and increased lipase (30.0%). CONCLUSION: The combination of INC280/buparlisib resulted in no clear activity in patients with recurrent PTEN-deficient glioblastoma. More stringent molecular selection strategies might produce better outcomes. TRIAL REGISTRATION: NCT01870726.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Aminopyridines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Benzamides , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Female , Follow-Up Studies , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Imidazoles/administration & dosage , Male , Maximum Tolerated Dose , Middle Aged , Morpholines/administration & dosage , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , PTEN Phosphohydrolase/genetics , Prognosis , Proto-Oncogene Proteins c-met/genetics , Tissue Distribution , Triazines/administration & dosage
8.
Cancer Sci ; 111(2): 536-547, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31778267

ABSTRACT

Capmatinib is an oral, ATP-competitive, and highly potent, type 1b MET inhibitor. Herein, we report phase 1 dose-escalation results for capmatinib in advanced MET-positive solid tumor patients and dose expansion in advanced non-lung tumors. Capmatinib was well tolerated with a manageable safety profile across all explored doses. Dose-limiting toxicities (DLT) occurred at 200 mg twice daily (bid), 250 mg bid, and 450 mg bid capsules; however, no DLT were reported at 600 mg bid (capsules). Capmatinib tablets at 400 mg bid had comparable tolerability and exposure to that of 600 mg bid capsules. Maximum tolerated dose was not reached; recommended phase 2 dose was 400 mg bid tablets/600 mg bid capsules; at this dose, Ctrough >EC90 (90% inhibition of c-MET phosphorylation in animal models) is expected to be achieved and maintained. Among the dose-expansion patients (N = 38), best overall response across all cohorts was stable disease (gastric cancer 22%, hepatocellular carcinoma 46%, other indications 28%); two other indication patients with gene copy number (GCN) ≥6 achieved substantial tumor reduction. Near-complete immunohistochemically determined phospho-MET inhibition (H-score = 2) was shown following capmatinib 450 mg bid capsule in paired biopsies obtained from one advanced colorectal cancer patient. Incidence of high-level MET GCN (GCN ≥6) and MET-overexpressing (immunohistochemistry 3+) tumors in the expansion cohorts was 8% and 13%, respectively; no MET mutations were observed. Thus, the recommended phase 2 dose (RP2D) of capmatinib was 600 mg bid capsule/400 mg bid tablet. Capmatinib was well tolerated and showed antitumor activity and acceptable safety profile at the RP2D. (ClinicalTrials.gov Identifier: NCT01324479).


Subject(s)
Imidazoles/administration & dosage , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Triazines/administration & dosage , Aged , Benzamides , Capsules , Dose-Response Relationship, Drug , Female , Humans , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Male , Middle Aged , Mutation , Neoplasms/metabolism , Phosphorylation , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Tablets , Treatment Outcome , Triazines/adverse effects , Triazines/pharmacokinetics
9.
Leukemia ; 33(12): 2924-2933, 2019 12.
Article in English | MEDLINE | ID: mdl-31092894

ABSTRACT

PIM447, a novel pan-PIM inhibitor, has shown preclinical activity in multiple myeloma (MM). In the multicenter, open-label, first-in-human study, patients with relapsed and/or refractory MM were enrolled to determine the maximum-tolerated dose (MTD) or recommended dose (RD), safety, pharmacokinetics, and preliminary anti-myeloma activity of PIM447. PIM447 was administered in escalating oral doses of 70-700 mg once daily (q.d.) for 28-day continuous cycles. Seventy-nine patients with a median of four prior therapies were enrolled. Seventy-seven patients (97.5%) had an adverse event (AE) suspected as treatment related, with treatment-related grade 3/4 AEs being mostly hematologic. Eleven dose-limiting toxicities occurred, and an MTD of 500 mg q.d. and an RD of 300 mg q.d. were established. The main reason for discontinuation was disease progression in 54 patients (68.4%). In the entire study population, a disease control rate of 72.2%, a clinical benefit rate of 25.3%, and an overall response rate of 8.9% were observed per modified International Myeloma Working Group criteria. Median progression-free survival at the RD was 10.9 months. PIM447 was well tolerated and demonstrated single-agent antitumor activity in relapsed/refractory MM patients, providing proof of principle for Pim (Proviral Insertions of Moloney Murine leukemia virus) kinase inhibition as a novel therapeutic approach in MM.


Subject(s)
Antineoplastic Agents/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Monitoring , Drug Resistance , Female , Humans , Male , Middle Aged , Multiple Myeloma/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Recurrence , Treatment Outcome
10.
Eur J Drug Metab Pharmacokinet ; 44(5): 669-680, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31030415

ABSTRACT

BACKGROUND AND OBJECTIVES: MET401 is a potent and selective c-Met inhibitor with a novel triazolopyrimidine scaffold. The aim of this study was to determine the pharmacokinetic profile of MET401 in preclinical species, and to identify the metabolic soft spot and enzyme involved, in order to help medicinal chemists to modify the compound to improve the pharmacokinetic profile. METHODS: A metabolite identification study was performed in different liver fractions from various species. Chemical inhibition with selective cytochrome P450 (CYP) and molybdenum hydroxylase inhibitors was carried out to identify the enzyme involved. The deuterium substitution strategy was adopted to reduce metabolism. Pharmacokinetic studies were performed in rats to confirm the effect. RESULTS: Although M-2 is a minor metabolite in liver microsomal incubations, it became the predominant metabolite in incubations with liver S9, cytosol, hepatocytes and rat pharmacokinetic study. M-2 was synthesized enzymatically and the structure was identified as a mono-oxidation on the triazolopyrimidine moiety. The M-2 formation was ascribed to aldehyde oxidase (AO)-mediated metabolism based on the following evidence-M-2 production was NADPH independent, pan-CYP inhibitor 1-aminobenzotriazole and xanthine oxidase inhibitor allopurinol did not inhibit M-2 formation, and AO inhibitors menadione and raloxifene inhibited M-2 formation. The deuterated analog MET763 demonstrated an improved pharmacokinetic profile with lower clearance, longer terminal half-life and double oral exposure compared with MET401 in rats. CONCLUSIONS: These results indicate that the main metabolic pathway of MET401 is AO-mediated metabolism, which leads to poor in vivo pharmacokinetic profiles in rodents. The deuterium substitution strategy could be used to reduce AO-mediated metabolism liability.


Subject(s)
Aldehyde Oxidase/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Cytochrome P-450 Enzyme System/metabolism , Cytosol/metabolism , Dogs , Female , Guinea Pigs , Hepatocytes/metabolism , Humans , Liver/metabolism , Macaca fascicularis , Male , Metabolic Networks and Pathways/physiology , Mice , Microsomes, Liver/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
11.
J Clin Oncol ; 36(31): 3101-3109, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30156984

ABSTRACT

PURPOSE: MET dysregulation occurs in up to 26% of non-small-cell lung cancers (NSCLCs) after epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI) treatment. Capmatinib (INC280) is a potent and selective MET inhibitor with preclinical activity in combination with gefitinib in EGFR-mutant, MET-amplified/overexpressing models of acquired EGFR-TKI resistance. This phase Ib/II study investigated the safety and efficacy of capmatinib plus gefitinib in patients with EGFR-mutated, MET-dysregulated (amplified/overexpressing) NSCLC who experienced disease progression while receiving EGFR-TKI treatment. METHODS: Patients in phase Ib received capmatinib 100- to 800-mg capsules once per day or 200- to 600-mg capsules or tablets twice per day, plus gefitinib 250 mg once per day. Patients in phase II received the recommended phase II dose. The primary end point was the overall response rate (ORR) per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. RESULTS: Sixty-one patients were treated in phase Ib, and 100 were treated in phase II. The recommended phase II dose was capmatinib 400 mg twice per day plus gefitinib 250 mg once per day. Preliminary clinical activity was observed, with an ORR across phase Ib/II of 27%. Increased activity was seen in patients with high MET-amplified tumors, with a phase II ORR of 47% in patients with a MET gene copy number ≥ 6. Across phases Ib and II, the most common drug-related adverse events were nausea (28%), peripheral edema (22%), decreased appetite (21%), and rash (20%); the most common drug-related grade 3/4 adverse events were increased amylase and lipase levels (both 6%). No significant drug-drug interactions between capmatinib and gefitinib were evident. CONCLUSION: This study, focused on a predominant EGFR-TKI resistance mechanism in patients with EGFR-mutated NSCLC, shows that the combination of capmatinib with gefitinib is a promising treatment for patients with EGFR-mutated, MET-dysregulated NSCLC, particularly MET-amplified disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Benzamides , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/genetics , Female , Gefitinib/administration & dosage , Gefitinib/adverse effects , Gefitinib/pharmacokinetics , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Mutation , Proto-Oncogene Proteins c-met/metabolism , Triazines/administration & dosage , Triazines/adverse effects , Triazines/pharmacokinetics
12.
J Appl Toxicol ; 36(2): 320-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26201057

ABSTRACT

Cytochrome P450 (CYP) induction is a key risk factor of clinical drug-drug interactions that has to be mitigated in the early phases of drug discovery. Three-dimensional (3D) cultures of hepatocytes in vitro have recently emerged as a potentially better platform to recapitulate the in vivo liver structure and to maintain long-term hepatic functions as compared with conventional two-dimensional (2D) monolayer cultures. However, the majority of published studies on 3D hepatocyte models use rat hepatocytes and the response to CYP inducers between rodents and humans is distinct. In the present study, we constructed tethered spheroids on RGD/galactose-conjugated membranes as an in vitro 3D model using cryopreserved human hepatocytes. CYP3A4 mRNA expression in the tethered spheroids was induced to a significantly greater extent than those in the collagen sandwich cultures, indicating the transcriptional regulation was more sensitive to the CYP inducers in the 3D model. Induction of CYP1A2, CYP2B6 and CYP3A4 activities in the tethered spheroids were comparable to, if not higher than that observed in the collagen sandwich cultures. The membrane-based model is readily integrated into multi-well plates for higher-throughput drug testing applications, which might be an alternative model to screen the CYP induction potential in vitro with more physiological relevance.


Subject(s)
Cells, Cultured/drug effects , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 Enzyme System/metabolism , Drug Evaluation/methods , Drug Interactions/physiology , Hepatocytes/drug effects , Humans
13.
Drug Metab Dispos ; 42(5): 839-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24595680

ABSTRACT

It has been proposed that in humans 4ß-hydroxycholesterol is formed mainly by CYP3A-catalyzed metabolism of cholesterol and thus may serve as an endogenous marker for CYP3A activity. The cynomolgus monkey is widely used as one of the nonrodent preclinical safety species in pharmaceutical research. In the current study, the potential application of 4ß-hydroxycholesterol as an endogenous biomarker of CYP3A in response to drug treatment was evaluated in cynomolgus monkeys. Following multiple oral administration of rifampicin (a known CYP3A inducer) at 15 mg/kg/d in cynomolgus monkeys, the mean serum 4ß-hydroxycholesterol levels increased 4-fold from the baseline of 55.3 ± 21.7 to 221 ± 53.4 ng/ml. The mean concentration ratios of 4ß-hydroxycholesterol to cholesterol increased 5-fold. The data suggest that 4ß-hydroxycholesterol formation from cholesterol metabolism was induced by rifampicin treatment in monkeys. This observation correlated with the metabolism of midazolam (a probe substrate of CYP3A activity) monitored in the same study. The serum exposure (area under the curve) of midazolam was markedly decreased by ∼95%, confirming the induction of CYP3A catalytic activity by rifampicin treatment in monkeys. The formation of 4ß-hydroxycholesterol from cholesterol was specifically mediated by recombinant cynomolgus CYP3A8 and CYP3A5. The Km values of CYP3A8 and CYP3A5 for 4ß-hydroxycholesterol formation from cholesterol were 204 and 104 µM, respectively, and Vmax values were 0.600 and 0.310 pg/pmol/min, respectively. The results suggest that 4ß-hydroxycholesterol can be used as an endogenous biomarker to identify strong CYP3A inducers in cynomolgus monkeys, which may help to evaluate drug-drug interaction potential of drug candidates in preclinical settings.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Hydroxycholesterols/blood , Administration, Oral , Animals , Biomarkers/blood , Biotransformation , Cholesterol/metabolism , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 CYP3A/genetics , Drug Evaluation, Preclinical , Enzyme Induction , Female , Macaca fascicularis , Male , Midazolam/blood , Midazolam/pharmacokinetics , Rifampin/pharmacology , Substrate Specificity , Tandem Mass Spectrometry
14.
Cancer Res ; 70(13): 5518-27, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20551065

ABSTRACT

The BRAF(V600E) mutation is common in several human cancers, especially melanoma. RG7204 (PLX4032) is a small-molecule inhibitor of BRAF(V600E) kinase activity that is in phase II and phase III clinical testing. Here, we report a preclinical characterization of the antitumor activity of RG7204 using established in vitro and in vivo models of malignant melanoma. RG7204 potently inhibited proliferation and mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase and ERK phosphorylation in a panel of tumor cell lines, including melanoma cell lines expressing BRAF(V600E) or other mutant BRAF proteins altered at codon 600. In contrast, RG7204 lacked activity in cell lines that express wild-type BRAF or non-V600 mutations. In several tumor xenograft models of BRAF(V600E)-expressing melanoma, we found that RG7204 treatment caused partial or complete tumor regressions and improved animal survival, in a dose-dependent manner. There was no toxicity observed in any dose group in any of the in vivo models tested. Our findings offer evidence of the potent antitumor activity of RG7204 against melanomas harboring the mutant BRAF(V600E) gene.


Subject(s)
Indoles/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Nude , Mutation , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL