Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Transl Sci ; 15(5): 1143-1154, 2022 05.
Article in English | MEDLINE | ID: mdl-35041775

ABSTRACT

The ALK in Lung Cancer Trial of brigAtinib in First Line (ALTA-1L) compared brigatinib versus crizotinib in anaplastic lymphoma kinase (ALK) inhibitor-naive patients with ALK+ non-small cell lung cancer (NSCLC). A population pharmacokinetic (PK) model was used to estimate brigatinib exposures for exposure-efficacy and exposure-safety analyses in ALTA-1L. A previously developed population PK model for brigatinib was applied to estimate brigatinib PK parameters. Relationships between static (time-independent) and dynamic (time-varying) exposure metrics and efficacy (progression-free survival [PFS], objective response rate [ORR], and intracranial ORR [iORR]) and safety outcomes (selected grade ≥2 and grade ≥3 adverse events [AEs]) were evaluated using logistic regression and time-to-event analyses. There were no meaningful differences in brigatinib PK in the first-line and second-line settings, supporting use of the previous population PK model for the first-line population. Exposure-response analyses showed no significant effect of time-varying brigatinib exposure on PFS. Brigatinib exposure was not significantly related to ORR, but higher exposure was associated with higher iORR (odds ratio: 1.13, 95% confidence interval: 1.01-1.28, p = 0.049). Across the observed median exposure (5th-95th percentile) at steady state for 180 mg once daily, the predicted probability of iORR was 0.83 (0.58-0.99). AEs significantly associated with higher exposure were elevated lipase (grade ≥3) and amylase (grade ≥2). Time to first brigatinib dose reduction was not related to exposure. These results support the benefit-risk profile of first-line brigatinib 180 mg once daily (7-day lead-in dose at 90 mg once daily) in patients with ALK+ NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Anaplastic Lymphoma Kinase , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Organophosphorus Compounds , Protein Kinase Inhibitors/adverse effects , Pyrimidines/adverse effects
2.
Clin Pharmacol Ther ; 105(5): 1234-1243, 2019 05.
Article in English | MEDLINE | ID: mdl-30347431

ABSTRACT

ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) is required for the production of ß-amyloid (Aß) peptides and is considered a potential treatment target for Alzheimer's disease (AD). To support Japan's participation in the global clinical development program, we characterized the safety, pharmacokinetics (PKs), and pharmacodynamics of the BACE1 inhibitor verubecestat (MK-8931) in 24 healthy Japanese adults in a two-part, single-center, randomized, placebo-controlled phase I trial (protocol MK-8931-007) and compared the results with historical data from non-Japanese subjects. Both single (20, 100, and 450 mg) and multiple (80 and 150 mg once daily for 14 days) doses of verubecestat were well tolerated. Verubecestat's PK profile was similar in Japanese and non-Japanese subjects. Verubecestat also reduced mean cerebrospinal fluid concentrations of the Aß proteins Aß40, Aß42, and soluble ß fragment of amyloid precursor protein; the level of reduction was comparable between Japanese and non-Japanese subjects. These results support the continued global development of verubecestat as a potential disease-modifying agent for Japanese and non-Japanese subjects who are at risk for developing AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cyclic S-Oxides , Thiadiazines , Adult , Alzheimer Disease/ethnology , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Protein Precursor/blood , Amyloid beta-Protein Precursor/metabolism , Cyclic S-Oxides/administration & dosage , Cyclic S-Oxides/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Monitoring/methods , Female , Healthy Volunteers , Humans , Japan , Male , Thiadiazines/administration & dosage , Thiadiazines/pharmacokinetics
3.
Sci Transl Med ; 8(363): 363ra150, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27807285

ABSTRACT

ß-Amyloid (Aß) peptides are thought to be critically involved in the etiology of Alzheimer's disease (AD). The aspartyl protease ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) is required for the production of Aß, and BACE1 inhibition is thus an attractive target for the treatment of AD. We show that verubecestat (MK-8931) is a potent, selective, structurally unique BACE1 inhibitor that reduced plasma, cerebrospinal fluid (CSF), and brain concentrations of Aß40, Aß42, and sAPPß (a direct product of BACE1 enzymatic activity) after acute and chronic administration to rats and monkeys. Chronic treatment of rats and monkeys with verubecestat achieved exposures >40-fold higher than those being tested in clinical trials in AD patients yet did not elicit many of the adverse effects previously attributed to BACE inhibition, such as reduced nerve myelination, neurodegeneration, altered glucose homeostasis, or hepatotoxicity. Fur hypopigmentation was observed in rabbits and mice but not in monkeys. Single and multiple doses were generally well tolerated and produced reductions in Aß40, Aß42, and sAPPß in the CSF of both healthy human subjects and AD patients. The human data were fit to an amyloid pathway model that provided insight into the Aß pools affected by BACE1 inhibition and guided the choice of doses for subsequent clinical trials.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Aspartic Acid Endopeptidases/antagonists & inhibitors , Central Nervous System/metabolism , Cyclic S-Oxides/pharmacology , Thiadiazines/pharmacology , Administration, Oral , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Catalytic Domain , Crystallography, X-Ray , Drug Design , Female , Glucose/metabolism , Macaca fascicularis , Magnetic Resonance Spectroscopy , Mice , Myelin Sheath/chemistry , Peptides/chemistry , Rabbits , Rats
4.
Br J Clin Pharmacol ; 72(3): 415-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21535448

ABSTRACT

AIMS: An integrated population pharmacokinetic-pharmacodynamic model was developed with the following aims: to simultaneously describe pharmacokinetic behaviour of sugammadex and rocuronium; to establish the pharmacokinetic-pharmacodynamic model for rocuronium-induced neuromuscular blockade and reversal by sugammadex; to evaluate covariate effects; and to explore, by simulation, typical covariate effects on reversal time. METHODS: Data (n= 446) from eight sugammadex clinical studies covering men, women, non-Asians, Asians, paediatrics, adults and the elderly, with various degrees of renal impairment, were used. Modelling and simulation techniques based on physiological principles were applied to capture rocuronium and sugammadex pharmacokinetics and pharmacodynamics and to identify and quantify covariate effects. RESULTS: Sugammadex pharmacokinetics were affected by renal function, bodyweight and race, and rocuronium pharmacokinetics were affected by age, renal function and race. Sevoflurane potentiated rocuronium-induced neuromuscular blockade. Posterior predictive checks and bootstrapping illustrated the accuracy and robustness of the model. External validation showed concordance between observed and predicted reversal times, but interindividual variability in reversal time was pronounced. Simulated reversal times in typical adults were 0.8, 1.5 and 1.4 min upon reversal with sugammadex 16 mg kg(-1) 3 min after rocuronium, sugammadex 4 mg kg(-1) during deep neuromuscular blockade and sugammadex 2 mg kg(-1) during moderate blockade, respectively. Simulations indicated that reversal times were faster in paediatric patients and slightly slower in elderly patients compared with adults. Renal function did not affect reversal time. CONCLUSIONS: Simulations of the therapeutic dosing regimens demonstrated limited impact of age, renal function and sevoflurane use, as predicted reversal time in typical subjects was always <2 min.


Subject(s)
Androstanols/pharmacology , Neuromuscular Blockade , Neuromuscular Nondepolarizing Agents/pharmacology , gamma-Cyclodextrins/pharmacology , gamma-Cyclodextrins/pharmacokinetics , Adolescent , Adult , Aged , Aged, 80 and over , Androstanols/pharmacokinetics , Clinical Trials as Topic , Drug Interactions , Female , Glomerular Filtration Rate/drug effects , Humans , Kidney/drug effects , Kidney/metabolism , Male , Middle Aged , Models, Neurological , Muscle Relaxation/drug effects , Neuromuscular Nondepolarizing Agents/pharmacokinetics , Rocuronium , Sugammadex , Young Adult
5.
J Psychopharmacol ; 24(11): 1689-96, 2010 Nov.
Article in English | MEDLINE | ID: mdl-19939872

ABSTRACT

An ideal drug for outpatient treatments under conscious sedation would have both sedative and analgesic properties. CB1/CB2 agonists are expected to have sedative, amnestic, analgesic and anti-emetic properties. The main objective of this first study in humans was to assess the sedative properties of intravenous Org 26828. In addition, pharmacokinetics, amnestic properties, postural stability, and behavioural and cardiovascular effects were studied. Midazolam intravenous 0.1 mg/kg and placebo were used as controls. The pharmacokinetic parameters (Cmax and AUC0-inf) of the main metabolite Org 26761 were proportional to dose. No effects were observed after doses up to 0.3 µg/kg of Org 26828. Dose-related effects were observed at higher doses. Although subjects reported subjective sedation after administration of Org 26828 at 3 and 6 µg/kg, the observed sedation was considerably less than after midazolam. Doses higher than the maximum tolerated dose of 1 µg/kg of Org 26828 caused unpleasant central nervous system effects (anxiety, paranoia, hallucinations). Therefore, Org 26828 is not suitable for providing sedation for outpatient surgical procedures.


Subject(s)
Conscious Sedation , Dronabinol/analogs & derivatives , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/pharmacokinetics , Midazolam/pharmacology , Midazolam/pharmacokinetics , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/drug effects , Adult , Ambulatory Surgical Procedures , Dose-Response Relationship, Drug , Dronabinol/adverse effects , Dronabinol/blood , Dronabinol/pharmacokinetics , Dronabinol/pharmacology , Humans , Hypnotics and Sedatives/adverse effects , Infusions, Intravenous , Injections, Intravenous , Male , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...