Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Eur J Clin Pharmacol ; 71(6): 663-672, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939707

ABSTRACT

PURPOSE: The study aimed to characterize the population pharmacokinetics of panobinostat, a pan-deacetylase inhibitor that has demonstrated efficacy in combination with bortezomib and dexamethasone in patients with multiple myeloma. METHODS: A nonlinear mixed-effect model was used to fit plasma panobinostat concentration-time data collected from patients across 14 phase 1 and phase 2 trials following either oral or intravenous (IV) administration. The model was used to estimate bioavailabilities of the two oral formulations and the effects of demographic and clinical covariates on the central volume of distribution and clearance of panobinostat. RESULTS: A total of 7834 samples from 581 patients were analyzed. Panobinostat pharmacokinetic parameters were best characterized by a three-compartment model with first-order absorption and elimination. Bioavailability was 21.4 %. Median clearance was 33.1 L/h. Interindividual variability in clearance was 74 %. For Caucasian patients of median age 61 years, area under the curve (AUC) decreased from 104 to 88 ng · h/mL as body surface area (BSA) increased from the first to third quartiles, 1.8 to 2.1 m(2). For Caucasian patients of median BSA 1.9 m(2), AUC decreased from 102 to 95 ng · h/mL as age increased from the first to third quartiles, 51 to 70 years. For patients of median BSA and median age, AUC ranged across the four race categories from 80 to 116 ng · h/mL. Covariate analysis showed no impact on panobinostat clearance and volume by patients' sex, tumor type, kidney function, liver markers, or coadministered medications. However, separate analyses of dedicated studies have demonstrated effects of liver impairment and CYP3A4 inhibition. CONCLUSIONS: Although covariate analyses revealed significant effects of body size, age, and race on panobinostat pharmacokinetics, these effects were minor compared to the interindividual variability and therefore not clinically relevant when dosing panobinostat in populations similar to those studied.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Hematologic Neoplasms/drug therapy , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacokinetics , Indoles/administration & dosage , Indoles/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Area Under Curve , Biological Availability , Female , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Middle Aged , Panobinostat , Young Adult
2.
J Clin Pharmacol ; 53(1): 14-20, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23400739

ABSTRACT

Dovitinib is an oral multitargeted kinase inhibitor with potent activity against receptors for vascular endothelial growth factor, platelet-derived growth factor, and basic fibroblast growth factor. Initial phase 1 to 2 studies of dovitinib using a continuous daily dosing schedule has shown that dovitinib exhibits a prolonged and overproportional increase in dose and exposure relationship above 400 mg/d. To address this, intermittent dosing schedules were explored using a model-based approach. A semi-mechanistic population pharmcokinetic/pharmacodynamic (PD) model was developed from 4 dovitinib phase 1 studies with daily dosing schedules. Autoinduction of cytochrome P450 1A (CYP1A) responsible for dovitinib metabolism was described using an indirect response model. Simulation of dovitinib plasma concentration profiles following 4 intermittent dosing schedules suggested that intermittent dosing could prevent prolonged drug accumulation. Based on the simulated plasma profiles, PD response, and patient compliance, a 5-days-on/2-days-off intermittent dosing schedule was selected for a phase 1 to 2 clinical study. The observed dovitinib plasma concentrations in this study confirmed the model predictions. Furthermore, dovitinib was well tolerated, and antitumor activity was observed as well in this new study. The 5-days-on/2-days-off dosing schedule is currently used in a dovitinib registration trial and other clinical trials.


Subject(s)
Benzimidazoles/administration & dosage , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Quinolones/administration & dosage , Benzimidazoles/blood , Benzimidazoles/pharmacokinetics , Drug Administration Schedule , Female , Humans , Male , Models, Biological , Neoplasms/metabolism , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Quinolones/blood , Quinolones/pharmacokinetics , Vascular Endothelial Growth Factor A/blood
3.
J Clin Pharmacol ; 51(11): 1580-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21712497

ABSTRACT

Nilotinib, a potent orally bioavailable BCR-ABL tyrosine kinase inhibitor, is currently available as a hard gelatin capsule that must be swallowed whole. For patients who may have difficulty swallowing the intact capsule, an alternative mode of administration is desirable. The authors compared the bioavailability of nilotinib from the following administrations in 48 healthy subjects: (1) 400 mg nilotinib given as two 200-mg nilotinib intact capsules; (2) contents of two 200-mg nilotinib capsules, each capsule dispersed in 1 teaspoon of nonfat plain yogurt; and (3) contents of two 200-mg nilotinib capsules, each capsule dispersed in 1 teaspoon of applesauce. Nilotinib absorption was modestly increased following the administration of nilotinib dispersed in yogurt. The geometric mean ratios (90% confidence intervals) for nilotinib C(max), AUC(0-tlast), and AUC(0-inf) were 1.31 (1.22-1.41), 1.11 (1.05-1.16), and 1.08 (1.02-1.15), respectively. Administration of nilotinib dispersed in applesauce showed equivalent bioavailability compared with administration of nilotinib as intact capsules. The geometric mean ratios (90% confidence intervals) for nilotinib C(max), AUC(0-tlast), and AUC(0-inf) were 0.95 (0.88-1.02), 0.99 (0.94-1.04), and 0.97 (0.90-1.03), respectively. Each treatment was well tolerated in the study subjects. The data support a feasible alternative method of nilotinib administration; each capsule's contents may be dispersed in 1 teaspoon of applesauce and taken immediately.


Subject(s)
Food-Drug Interactions , Malus , Pyrimidines/pharmacokinetics , Yogurt , Administration, Oral , Area Under Curve , Biological Availability , Capsules/administration & dosage , Confidence Intervals , Cross-Over Studies , Deglutition Disorders , Female , Humans , Male , Middle Aged , Pyrimidines/administration & dosage , Therapeutic Equivalency
4.
Br J Clin Pharmacol ; 71(2): 199-206, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21219400

ABSTRACT

AIMS: The major objective of the present study was to investigate the effect of imatinib on the pharmacokinetics of paracetamol in patients with chronic myelogenous leukaemia (CML). METHODS: Patients (n = 12) received a single oral dose of acetaminophen 1000 mg on day 1 (control). On days 2-8, imatinib 400 mg was administered daily. On day 8 (treatment), another 1000 mg dose of paracetamol was administered 1 h after the morning dose of imatinib 400 mg. Blood and urine samples were collected for bioanalytical analyses. RESULTS: The area under the plasma concentration-time curve (AUC) for paracetamol, paracetamol glucuronide and paracetamol sulphate under control conditions was similar to that after treatment with imatinib; the 90% confidence interval of the log AUC ratio was within 0.8 to 1.25. Urinary excretion of paracetamol, paracetamol glucuronide and paracetamol sulphate was also unaffected by imatinib. The pharmacokinetics of paracetamol and imatinib in Korean patients with CML were similar to previous pharmacokinetic results in white patients with CML. Co-administration of a single dose of paracetamol and multiple doses of imatinib was well tolerated and safety profiles were similar to those of either drug alone. CONCLUSIONS: The pharmacokinetics of paracetamol and its major metabolites in the presence of imatinib were similar to those of the control conditions and the combination was well tolerated. These findings suggest that imatinib can be safely administered with paracetamol without dose adjustment of either drug.


Subject(s)
Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Antineoplastic Agents/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Acetaminophen/administration & dosage , Adult , Aged , Analgesics, Non-Narcotic/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Benzamides , Drug Administration Schedule , Drug Interactions , Drug Monitoring/methods , Female , Half-Life , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Middle Aged , Piperazines/administration & dosage , Piperazines/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/adverse effects
5.
J Clin Pharmacol ; 51(1): 75-83, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20702754

ABSTRACT

Nilotinib (Tasigna), an orally bioavailable second-generation BCR-ABL tyrosine kinase inhibitor, is approved for use in patients with chronic myeloid leukemia in chronic phase and accelerated phase who are resistant or intolerant to prior therapy, including imatinib. Previous in vitro studies indicated that nilotinib metabolism is primarily mediated by CYP3A4. To investigate the effect of CYP3A4 induction and inhibition on nilotinib pharmacokinetics, 2 studies were conducted in healthy volunteers prior to and following treatment with a strong inducer (rifampin) or inhibitor (ketoconazole). In the induction study, administration of rifampin 600 mg once daily for 8 days significantly increased urinary 6ß-hydroxycortisol/ cortisol ratio, from a preinduction baseline of 5.8 ± 2.7 to 18.0 ± 10.2 after 8 days of rifampin treatment, confirming an inductive effect on CYP3A4. Nilotinib oral clearance was increased by 4.8-fold, and the maximum serum concentration (C(max)) and area under the serum concentration-time curve (AUC) were decreased by 64% and 80%, respectively, in the induced state compared with baseline. In the inhibition study, ketoconazole 400 mg once daily for 6 days increased the C(max) and AUC of nilotinib by 1.8- and 3-fold, respectively, compared with nilotinib alone. These results indicate that concurrent use of strong CYP3A4 inducers or inhibitors may necessitate dosage adjustments of nilotinib and should be avoided when possible.


Subject(s)
Cytochrome P-450 CYP3A/drug effects , Ketoconazole/pharmacology , Pyrimidines/pharmacokinetics , Rifampin/pharmacology , Adult , Area Under Curve , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Female , Humans , Hydrocortisone/analogs & derivatives , Hydrocortisone/urine , Male , Young Adult
6.
Clin Drug Investig ; 31(3): 169-79, 2011.
Article in English | MEDLINE | ID: mdl-21184622

ABSTRACT

BACKGROUND AND OBJECTIVE: Nilotinib (Tasigna®), a highly selective and potent BCR-ABL tyrosine kinase inhibitor, is approved for the treatment of chronic myeloid leukaemia in the chronic phase (CML-CP) and the accelerated phase (CML-AP) in patients resistant or intolerant to prior therapy, including imatinib. Nilotinib has shown competitive inhibition of cytochrome P450 enzyme (CYP) 2C9 in vitro, but its effect on CYP2C9 activity in humans is unknown. This study evaluated the effects of nilotinib on the pharmacokinetics and pharmacodynamics of warfarin, a sensitive CYP2C9 substrate, in healthy subjects. METHODS: Twenty-four subjects (six female, 18 male, aged 21-65 years) were enrolled to receive a single oral dose of warfarin 25 mg with either a single oral dose of nilotinib 800 mg or matching placebo (all administered 30 minutes after consumption of a high-fat meal) in a crossover design. Serial blood samples were collected post-dose for determining serum concentrations of nilotinib and plasma concentrations of S- and R-warfarin. Prothrombin time (PT) and international normalized ratio (INR) values were determined as pharmacodynamic measures of warfarin activity. CYP2C9 genotyping was performed in all subjects using TaqMan® assay. RESULTS: Sixteen subjects were identified as CYP2C9 extensive metabolizers (EMs) and eight as intermediate metabolizers (IMs). There were no CYP2C9 poor metabolizers. Pharmacokinetic parameters of S- and R-warfarin were similar between the two treatments (warfarin + nilotinib vs warfarin alone) in both the EM and the IM groups. The geometric mean ratios (90% CIs) for the maximum concentration in plasma (C(max)) and area under the concentration-time curve from time zero to infinity (AUC(∞)) of S-warfarin in plasma in all subjects were 0.98 (0.95, 1.02) and 1.03 (0.99, 1.07), respectively, and for R-warfarin 1.00 (0.96, 1.04) and 1.02 (0.99, 1.04), respectively. Mean ratios for the maximum observed value and AUC from time zero to the last sampling time for PT were 1.00 (0.96, 1.04) and 1.00 (0.98, 1.02), respectively, and for the maximum observed value for INR and the AUC from time zero to the last sampling time for INR were 1.00 (0.97, 1.03) and 1.00 (0.99, 1.01), respectively. Mean ± SD serum nilotinib C(max) was 1872 ± 560 ng/mL, which is comparable to steady-state C(max) in CML and gastrointestinal stromal tumour patients receiving twice-daily 400 mg doses. Adverse events observed following either treatment were generally consistent with the known safety profiles of both drugs, and no new safety issues were observed. CONCLUSION: The study results demonstrate that nilotinib has no effect on single-dose warfarin pharmacokinetics and pharmacodynamics. This implies that nilotinib is unlikely to inhibit CYP2C9 activity in human subjects. These findings suggest that warfarin and nilotinib may be used concurrently as needed.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Warfarin/pharmacokinetics , Administration, Oral , Adult , Aged , Anticoagulants/adverse effects , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Area Under Curve , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Cross-Over Studies , Cytochrome P-450 CYP2C9 , Drug Interactions , Female , Fusion Proteins, bcr-abl/antagonists & inhibitors , Genotype , Humans , International Normalized Ratio , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Prothrombin Time , Pyrimidines/adverse effects , Single-Blind Method , Warfarin/adverse effects , Warfarin/pharmacology , Young Adult
7.
J Clin Pharmacol ; 50(8): 960-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20498287

ABSTRACT

Nilotinib (Tasigna), a highly selective and potent BCR-ABL tyrosine kinase inhibitor (TKI), is administered orally and has pH-dependent aqueous solubility, with lower dissolution at higher pH. This study evaluated the effect of esomeprazole on the pharmacokinetics of nilotinib in healthy participants. Twenty-two participants (6 women, 16 men, mean age of 44.9 +/- 12.9 years) were enrolled to receive nilotinib as a single oral 400-mg dose on days 1 and 13 and esomeprazole as 40 mg once daily on days 8 to 13. Serial blood samples were collected up to 72 hours after nilotinib dosing, and nilotinib serum concentrations were determined by a validated liquid chromatography/tandem mass spectrometry assay. Gastric pH was also monitored in all participants. When coadministered with esomeprazole, nilotinib C(max) was decreased by 27% and AUC(0-infinity) decreased by 34%. Nilotinib t(max) was prolonged from 4.0 to 6.0 hours, but t(1/2) was not altered. Mean gastric pH was 1.0 +/- 0.5 at baseline and increased to 2.79 +/- 2.50, 3.98 +/- 2.27, 5.30 +/- 1.70, 5.38 +/- 1.26, and 5.31 +/- 1.42 at predose and 1, 2, 3, and 4 hours after the fifth esomeprazole dose, respectively. These results suggested a modest reduction in the rate and extent of nilotinib absorption by esomeprazole. Nilotinib is a TKI that may be used concurrently with esomeprazole or other proton pump inhibitors.


Subject(s)
Esomeprazole/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proton Pump Inhibitors/pharmacology , Pyrimidines/pharmacokinetics , Adult , Area Under Curve , Female , Half-Life , Humans , Hydrogen-Ion Concentration , Male , Metabolic Clearance Rate , Middle Aged , Pyrimidines/adverse effects
8.
J Clin Pharmacol ; 50(2): 188-94, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19948946

ABSTRACT

Nilotinib (Tasigna; Novartis Pharmaceuticals) is a second-generation BCR-ABL tyrosine kinase inhibitor newly approved for the treatment of imatinib-resistant or imatinib-intolerant Philadelphia chromosome positive (Ph+) chronic myeloid leukemia in chronic phase or accelerated phase. This study evaluated the effect of grapefruit juice on the pharmacokinetics of nilotinib in 21 healthy male participants. All participants underwent 2 study periods during which they received a single oral dose of 400 mg nilotinib with 240 mL double-strength grapefruit juice or 240 mL water in a crossover fashion. Serial blood samples were collected for the determination of serum nilotinib concentrations by a validated liquid chromatography/tandem mass spectrometry assay. Concurrent intake of grapefruit juice increased the nilotinib peak concentration (C(max)) by 60% and the area under the serum concentration-time curve (AUC(0-infinity)) by 29% but did not affect the time to reach C(max) or the elimination half-life of nilotinib. The most common adverse events were headache and vomiting, which were mild or moderate in severity, and their frequency appeared to be similar between 2 treatments. Based on the currently available information about nilotinib and the observed extent of increase in nilotinib exposure, concurrent administration of nilotinib with grapefruit juice is not recommended.


Subject(s)
Citrus paradisi , Food-Drug Interactions , Pyrimidines/pharmacokinetics , Adolescent , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Beverages , Cross-Over Studies , Fusion Proteins, bcr-abl/metabolism , Headache/chemically induced , Humans , Male , Middle Aged , Protein-Tyrosine Kinases/metabolism , Pyrimidines/adverse effects , Vomiting/chemically induced , Young Adult
9.
Ther Drug Monit ; 31(5): 579-84, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19730279

ABSTRACT

BACKGROUND: Correlation analyses have demonstrated that maintaining an adequate imatinib (IM) trough concentration would be important for clinical response in patients with chronic myeloid leukemia (CML) and Kit-positive gastrointestinal stromal tumors. The objectives of the current work were to use a pharmacokinetic model to refine the trough levels obtained at different sampling times and to propose a therapeutic drug monitoring algorithm and an acceptable sampling time window for imatinib trough sampling. METHODS: The pharmacokinetics of IM in patients (pts) with CML were characterized based on historical data from a Phase III study. In the elimination phase the concentration of IM (C(t)) follows a mono-exponential decline, and the standardized trough concentration (C(min,std) = C(tau)) can be described by a simple algorithm C(min,std) = C(t)* exp(k(e) x Delta t), where Delta t = t - tau, and tau is 24 hours for qd or 12 hours for bid dosing and k(e) is the elimination rate constant. The percent deviation of C(t) from C(min,std) was simulated for different Delta t and k(e) values to define a sampling time window Delta t, within which the percent deviation is <20%. RESULTS: Simulation analysis shows that C(t) is largely dependent on Delta t and k(e). The percent deviation of C(t) at 3 hours before or after tau from C(min,std) will be 7.1%, 13.1%, and 23.4% for pts with low, typical, and high k(e) values, 0.023/hour, 0.041/hour, and 0.070/hour, respectively. However, if a correction is made for C(t) by the algorithm using the typical k(e) value of 0.041 per hour, the percent deviation at 3 hours will be reduced to 5.3%, 0%, and 9.1% for pts with low, typical, and high k(e) values, respectively. Even if the sampling window is extended to +/-6 hours, the corresponding percent deviation will still be reasonable: 10.2%, 0%, and 19.0%, respectively. CONCLUSION: By using the algorithm, the pharmacokinetic sampling window can be extended to a wider window to make the trough sampling easy to implement in the clinical setting, provided that the sampling time and dosing time are accurately recorded.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Algorithms , Benzamides , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Approval , Drug Monitoring , Humans , Imatinib Mesylate , Immunosuppressive Agents/pharmacology , Kidney Transplantation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors
10.
Clin Ther ; 31 Pt 2: 2459-69, 2009.
Article in English | MEDLINE | ID: mdl-20110053

ABSTRACT

BACKGROUND: Nilotinib is a second-generation BCR-ABL tyrosine kinase inhibitor approved for the treatment of patients who have imatinib-resistant Philadelphia chromosome-positive chronic myeloid leukemia in the chronic or accelerated phase or who are unable to tolerate imatinib. Nilotinib is metabolized in the liver via oxidation and hydroxylation pathways, mediated primarily by the cytochrome P450 3A4 isozyme. Interpatient variability in systemic exposure to nilotinib has been reported to range from 32% to 64%. OBJECTIVE: This study compared the pharmacokinetics of nilotinib in subjects with hepatic impairment and subjects with normal hepatic function. METHODS: Hepatic impairment was classified as mild (Child-Pugh grade A), moderate (Child-Pugh grade B), or severe (Child-Pugh grade C). Healthy control subjects were matched with hepatically impaired subjects by age (+/-10 years) and body weight (+/-20%). All subjects received a single oral dose of nilotinib 200 mg under fasted conditions, and serial blood samples were collected at specific times up to 120 hours after dosing. Serum nilotinib concentrations were measured using a validated LC-MS/MS assay with a lower limit of quantification of 2.5 ng/mL. The pharmacokinetic parameters analyzed were C(max), T(max), AUC(0-last), AUC(0-infinity), t(1/2), CL/F, and Vz/F. Tolerability assessments included adverse events (AEs), regular monitoring of clinical laboratory measures (eg, hematology, blood chemistry, urinalysis), physical examinations, vital signs, and ECGs. Each AE was evaluated in terms of its clinical significance, severity, duration, relation to study drug, and action taken. RESULTS: The study enrolled 18 subjects with hepatic impairment (all male; age range, 47-67 years; weight range, 73.9-103.9 kg) and 9 healthy controls (all male; age range, 36-62 years; weight range, 73.3-109.5 kg). Among subjects with hepatic impairment, 6 had mild impairment, 6 moderate impairment, and 6 severe impairment. The nilotinib AUC(0-infinity) was a mean of 35%, 35%, and 19% higher in subjects with mild, moderate, and severe impairment, respectively, compared with healthy controls. The nilotinib CL/F was lower in all hepatic-impairment groups compared with healthy controls. The mean (SD) t(1/2) was 15.1 (4.97) and 16.0 (9.13) hours in the mild-impairment and control groups, respectively, but was 21.6 (7.77) and 32.4 (10.7) hours in the moderate- and severe-impairment groups, respectively, reflecting the decrease in CL/F and/or increase in Vz/F in the latter 2 groups. All AEs were mild or moderate, and the frequency of AEs was not associated with the degree of hepatic impairment. AEs included abdominal pain (1 subject with mild impairment), dyspepsia (2 with mild impairment), flatulence (1 with severe impairment), nausea (1 with mild impairment), urinary tract infection (1 with mild impairment), back pain (1 each with mild impairment and severe impairment, 1 control subject), and headache (1 each with mild impairment and severe impairment). CONCLUSIONS: After a single 200-mg dose, nilotinib pharmacokinetics were modestly affected by hepatic impairment. The extent of change in nilotinib exposure in subjects with hepatic impairment was generally within the range of variability that has been observed clinically. The results of this study suggest that dose adjustment may not be necessary in patients with hepatic impairment. Nilotinib should be used with caution, and careful clinical monitoring is recommended in this population. ClinicalTrials.gov identifier: NCT00418626.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Hepatic Insufficiency/physiopathology , Pyrimidines/pharmacokinetics , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Area Under Curve , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Male , Middle Aged , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/adverse effects , Pyrimidines/blood
11.
Clin Pharmacokinet ; 47(12): 807-16, 2008.
Article in English | MEDLINE | ID: mdl-19026036

ABSTRACT

BACKGROUND AND OBJECTIVE: Midostaurin, a novel potent inhibitor of protein kinase C enzyme and class III receptor tyrosine kinases, including Fms-like tyrosine kinase-3 (FLT3) and c-KIT, shows time-dependent pharmacokinetics in human subjects, presumably due to enzyme auto-induction. The purpose of this study was to develop a mechanism-based population pharmacokinetic model to describe the plasma concentration profiles of midostaurin and its metabolites and to characterize the time course of auto-induction. SUBJECTS AND METHODS: Data from 37 diabetic patients who received oral doses of midostaurin (25 mg twice daily, 50 mg twice daily or 75 mg twice daily) for 28 days were analysed using nonlinear mixed-effects modelling. The structural model included a gut compartment for drug input and central and peripheral compartments for midostaurin, with drug output from the central compartment to either of two compartments for the midostaurin metabolites CGP62221 and CGP52421. Different enzyme induction sub-models were evaluated to account for the observed time-dependent decrease in midostaurin concentrations. RESULTS: An enzyme turnover model, with CGP62221 formation (CL(1)) being a linear process but CGP52421 formation (CL(2)) being inducible, was found to be most appropriate. In the pre-induced state, CL(1) and CL(2) of midostaurin were determined to be 1.47 L/h and 0.501 L/h, respectively. At the end of 28 days of dosing, CL(2) was increased by 5.2-, 6.6- and 6.9-fold in the 25 mg, 50 mg and 75 mg groups, respectively, resulting in a 2.1- to 2.5-fold increase in total clearance of midostaurin. The final model estimated a mean maximum fold of induction (E(max)) of 8.61 and a concentration producing 50% of the E(max) (EC(50)) of 1700 ng/mL (approximately 2.9 micromol/L) for CGP52421-mediated enzyme induction. CONCLUSIONS: The population pharmacokinetic model that was developed was able to describe the time-dependent pharmacokinetic profiles of midostaurin and its auto-induction mechanism. Thus it may be useful for designing an appropriate dosage regimen for midostaurin. The unique feature of this model included a precursor compartment that was able to capture the time delays of auto-induction. The use of such precursor extension in the model may be applicable to other drugs showing long time delays in enzyme auto-induction.


Subject(s)
Models, Biological , Staurosporine/analogs & derivatives , Administration, Oral , Aged , Algorithms , Chromatography, High Pressure Liquid , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Male , Middle Aged , Protein Kinase C/antagonists & inhibitors , Staurosporine/administration & dosage , Staurosporine/metabolism , Staurosporine/pharmacokinetics , Time Factors
13.
Drug Metab Dispos ; 36(10): 2043-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18625688

ABSTRACT

The bisphosphonate zoledronic acid is a potent inhibitor of osteoclast-mediated bone resorption. To investigate drug biodistribution and elimination, (14)C-zoledronic acid was administered intravenously to rats and dogs in single or multiple doses and assessed for its in vitro blood distribution and plasma protein binding in rat, dog, and human. Drug exposure in plasma, bones, and noncalcified tissues was investigated up to 240 days in rats and 96 h in dogs using radiometry after dissection. Drug biodistribution in the rat and within selected bones from dog was assessed by autoradiography. Concentrations of radioactivity showed a rapid decline in plasma and noncalcified tissue but only a slow decline in bone, to approximately 50% of peak at 240 days post dose, whereas the terminal half-lives (50-200 days) were similar in bone and noncalcified tissues, suggesting redistribution of drug from the former rather than prolonged retention in the latter. Uptake was highest in cancellous bone and axial skeleton. At 96 h after dose, the fraction of dose excreted was 36% in rat and 60% in dog; 94 to 96% of the excreted radioactivity was found in urine. Blood/plasma concentration ratios were 0.52 to 0.59, and plasma protein binding of zoledronic acid was moderate to low in all species. The results suggest that a fraction of zoledronic acid is reversibly taken up by the skeleton, the elimination of drug is mainly by renal excretion, and the disposition in blood and noncalcified tissue is governed by extensive uptake into and slow release from bone.


Subject(s)
Blood Proteins/metabolism , Bone Density Conservation Agents/pharmacokinetics , Diphosphonates/pharmacokinetics , Imidazoles/pharmacokinetics , Animals , Area Under Curve , Autoradiography , Bone Density Conservation Agents/metabolism , Bone Density Conservation Agents/urine , Diphosphonates/metabolism , Diphosphonates/urine , Dogs , Feces , Imidazoles/metabolism , Imidazoles/urine , Male , Protein Binding , Rats , Sensitivity and Specificity , Tissue Distribution , Zoledronic Acid
14.
J Clin Pharmacol ; 48(6): 763-75, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18508951

ABSTRACT

Midostaurin is a novel potent inhibitor of both protein kinase C and the major receptor for vascular endothelial growth factor involved in angiogenesis, presenting a rationale for its use in diabetic retinopathy. This study evaluated the safety and pharmacokinetics of midostaurin following multiple oral doses of midostaurin for 28 days at 4 dose levels (25 mg bid, 50 mg bid, 75 mg bid, 75 mg tid), as well as a single oral 100-mg dose in patients with diabetes mellitus (n = 9-13 per dose cohort). Pharmacokinetic parameters were determined on days 1 and 28 based on the plasma concentrations of midostaurin and its metabolites, CGP62221 and CGP52421. The plasma exposures (C(max) and AUC(0-tau)) of midostaurin and metabolites increased less than proportionally over the dose range of 25 to 100 mg, showing a 2.2-fold increase after the first dose. Midostaurin concentrations increased during the first 3 to 6 days of dosing, then declined with time (by 30%-50%) until a steady state was achieved, representing an average accumulation factor (R) of 1.7. CGP62221 showed a similar concentration-time pattern as midostaurin (R = 2.5), but CGP52421 accumulated significantly (R = 18.8). A high-fat meal was found to significantly increase the C(max) and AUC(0-12 h) of midostaurin by 1.5-fold (P = .04) and 1.8-fold (P = .01), respectively, compared with taking the drug after an overnight fast. Midostaurin administered at 50 to 225 mg/day appeared to be generally safe in this group of patients. The most common treatment-related adverse events (eg, loose stools, nausea, vomiting, and headache) were found to be dose related, and the frequency increased markedly above the 150-mg/day dose level.


Subject(s)
Diabetes Mellitus/drug therapy , Enzyme Inhibitors/pharmacokinetics , Staurosporine/analogs & derivatives , Adult , Aged , Area Under Curve , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Female , Humans , Male , Middle Aged , Protein Kinase C/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Staurosporine/administration & dosage , Staurosporine/adverse effects , Staurosporine/pharmacokinetics , Time Factors
15.
BMC Clin Pharmacol ; 8: 2, 2008 Mar 31.
Article in English | MEDLINE | ID: mdl-18377658

ABSTRACT

BACKGROUND: Cases of impaired renal function have been reported in patients who had been treated with both zoledronic acid and thalidomide for myeloma. Hence, we conducted a safety study of zoledronic acid and thalidomide in myeloma patients participating in a trial of maintenance therapy. METHODS: Twenty-four patients who were enrolled in a large randomized trial of thalidomide vs no thalidomide maintenance therapy for myeloma, in which all patients also received zoledronic acid, were recruited to a pharmacokinetic and renal safety sub-study, and followed for up to 16 months. RESULTS: No significant differences by Wilcoxon rank-sum statistic were found in zoledronic acid pharmacokinetics or renal safety for up to 16 months in patients randomized to thalidomide or not. CONCLUSION: In myeloma patients receiving maintenance therapy, the combination of zoledronic acid and thalidomide appears to confer no additional renal safety risks over zoledronic acid alone.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Diphosphonates/pharmacokinetics , Imidazoles/pharmacokinetics , Kidney/drug effects , Multiple Myeloma/drug therapy , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Creatinine/blood , Diphosphonates/administration & dosage , Diphosphonates/blood , Female , Humans , Imidazoles/administration & dosage , Imidazoles/blood , Male , Middle Aged , Multiple Myeloma/blood , Prednisolone/administration & dosage , Thalidomide/administration & dosage , Zoledronic Acid
16.
Br J Clin Pharmacol ; 65(6): 885-92, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18384443

ABSTRACT

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: Imatinib, a tyrosine kinase inhibitor, exhibits a competitive inhibition on the CYP450 2D6 isozyme with a K(i) value of 7.5 microm. However, the clinical significance of the inhibition and its relevance to 2D6 polymorphisms have not been evaluated. The pharmacokinetics of imatinib have been well studied in Caucasians, but not in a Chinese population. Metoprolol, a CYP2D6 substrate, has different clearances among patients with different CYP2D6 genotypes. It is often used as a CYP2D6 probe substrate for clinical drug-drug interaction studies. WHAT THIS STUDY ADDS: Co-administration of imatinib at 400 mg twice daily increased the plasma AUC of metoprolol by approximately 23% in 20 Chinese patients with chronic myeloid leukaemia (CML), about 17% increase in CYP2D6 intermediate metabolizers (IMs) (n = 6), 24% in extensive metabolizers (EMs) (n = 13), and 28% for the subject with unknown 2D6 status (n = 1) suggesting that imatinib has a weak to moderate inhibition on CYP2D6 in vivo. * The clearance of imatinib in Chinese patients with CML showed no difference between CYP2D6 IMs and EMs, and no major difference from Caucasian patients with CML based on data reported in the literature. AIMS To investigate the effect of imatinib on the pharmacokinetics of a CYP2D6 substrate, metoprolol, in patients with chronic myeloid leukaemia (CML). The pharmacokinetics of imatinib were also studied in these patients. METHODS: Patients (n = 20) received a single oral dose of metoprolol 100 mg on day 1 after an overnight fast. On days 2-10, imatinib 400 mg was administered twice daily. On day 8, another 100 mg dose of metoprolol was administered 1 h after the morning dose of imatinib 400 mg. Blood samples for metoprolol and alpha-hydroxymetoprolol measurement were taken on study days 1 and 8, and on day 8 for imatinib. RESULTS: Of the 20 patients enrolled, six patients (30%) were CYP2D6 intermediate metabolizers (IMs), 13 (65%) extensive metabolizers (EMs), and the CYP2D6 status in one patient was unknown. In the presence of 400 mg twice daily imatinib, the mean metoprolol AUC was increased by 17% in IMs (from 1190 to 1390 ng ml(-1) h), and 24% in EMs (from 660 to 818 ng ml(-1) h). Patients classified as CYP2D6 IMs had an approximately 1.8-fold higher plasma metoprolol exposure than those classified as EMs. The oral clearance of imatinib was 11.0 +/- 2.0 l h(-1) and 11.8 +/- 4.1 l h(-1) for CYP2D6 IMs and EMs, respectively. CONCLUSIONS: Co-administration of a high dose of imatinib resulted in a small or moderate increase in metoprolol plasma exposure in all patients regardless of CYP2D6 status. The clearance of imatinib showed no difference between CYP2D6 IMs and EMs.


Subject(s)
Adrenergic beta-Antagonists/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Metoprolol/pharmacokinetics , Piperazines/pharmacology , Pyrimidines/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Adult , Antineoplastic Agents/therapeutic use , Asian People/genetics , Benzamides , Cytochrome P-450 CYP2D6/genetics , Drug Interactions , Female , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Male , Metoprolol/therapeutic use , Middle Aged , Piperazines/therapeutic use , Polymorphism, Genetic , Pyrimidines/therapeutic use , Statistics as Topic
17.
Clin Pharmacokinet ; 44(9): 879-94, 2005.
Article in English | MEDLINE | ID: mdl-16122278

ABSTRACT

Imatinib is a potent and selective inhibitor of the protein tyrosine kinase Bcr-Abl, platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta) and KIT. Imatinib is approved for the treatment of chronic myeloid leukaemia (CML) and gastrointestinal stromal tumour (GIST), which have dysregulated activity of an imatinib-sensitive kinase as the underlying pathogenetic feature. Pharmacokinetic studies of imatinib in healthy volunteers and patients with CML, GIST and other cancers show that orally administered imatinib is well absorbed, and has an absolute bioavailability of 98% irrespective of oral dosage form (solution, capsule, tablet) or dosage strength (100 mg, 400 mg). Food has no relevant impact on the rate or extent of bioavailability. The terminal elimination half-life is approximately 18 hours. Imatinib plasma concentrations predictably increase by 2- to 3-fold when reaching steady state with 400mg once-daily administration, to 2.6 +/- 0.8 microg/mL at peak and 1.2 +/- 0.8 microg/mL at trough, exceeding the 0.5 microg/mL (1 micromol/L) concentrations needed for tyrosine kinase inhibition in vitro and leading to normalisation of haematological parameters in the large majority of patients with CML irrespective of baseline white blood cell count. Imatinib is approximately 95% bound to human plasma proteins, mainly albumin and alpha1-acid glycoprotein. The drug is eliminated predominantly via the bile in the form of metabolites, one of which (CGP 74588) shows comparable pharmacological activity to the parent drug. The faecal to urinary excretion ratio is approximately 5:1. Imatinib is metabolised mainly by the cytochrome P450 (CYP) 3A4 or CYP3A5 and can competitively inhibit the metabolism of drugs that are CYP3A4 or CYP3A5 substrates. Interactions may occur between imatinib and inhibitors or inducers of these enzymes, leading to changes in the plasma concentration of imatinib as well as coadministered drugs. Hepatic and renal dysfunction, and the presence of liver metastases, may result in more variable and increased exposure to the drug, although typically not necessitating dosage adjustment. Age (range 18-70 years), race, sex and bodyweight do not appreciably impact the pharmacokinetics of imatinib.


Subject(s)
Piperazines/pharmacokinetics , Pyrimidines/pharmacokinetics , Absorption , Animals , Benzamides , Drug Interactions , Humans , Imatinib Mesylate , Metabolic Clearance Rate , Microdialysis , Tissue Distribution
18.
Clin Pharmacol Ther ; 78(1): 69-80, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16003295

ABSTRACT

OBJECTIVE: Acromegaly is a serious hormonal disorder resulting from a pituitary adenoma causing excess growth hormone (GH) production. Somatostatin analogs such as octreotide have been the medical treatment of choice. SOM230, a novel somatostatin analog, was compared with octreotide with respect to pharmacokinetic (PK) profiles and inhibition of GH secretion in acromegalic patients. METHODS: In a double-blind, 3-period, crossover, proof-of-concept study, 12 patients with active acromegaly were randomized to single subcutaneous doses of SOM230 (100 and 250 microg) and octreotide (100 microg). Concentrations of SOM230, octreotide, and GH were determined at designated times after dosing and at baseline. The PK properties of SOM230 and octreotide and the relationship of PK with GH were investigated by a nonlinear mixed-effects modeling analysis. RESULTS: The apparent clearance for SOM230 is approximately half of that for octreotide (8.0 L/h versus 15.8 L/h). The elimination half-life for SOM230 is about 5 times longer than that for octreotide (11.8 hours versus 2.3 hours). The relationship between GH levels and plasma concentrations of SOM230 and octreotide is well described by a direct inhibitory model. The test drug concentration level at which half of the maximum drug effect is observed (EC50) is 46 and 553 pg/mL for octreotide and SOM230, respectively, with large interpatient variability (coefficients of variation, 164% and 65%, respectively), mainly attributable to the heterogeneous responses among patients. CONCLUSIONS: SOM230 demonstrates a lower clearance and longer half-life than octreotide, which compensates for the lower potency in GH inhibition. As a result of the lower interpatient variability for EC50 , SOM230 is expected to have a more uniform clinical GH inhibition than octreotide for acromegalic patients at a clinically effective dosing regimen.


Subject(s)
Acromegaly/drug therapy , Octreotide/pharmacology , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Acromegaly/blood , Acromegaly/diagnosis , Adult , Age Factors , Algorithms , Body Weight , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Female , Human Growth Hormone/antagonists & inhibitors , Human Growth Hormone/blood , Humans , Injections, Subcutaneous , Luminescent Measurements , Male , Middle Aged , Models, Biological , Octreotide/blood , Octreotide/therapeutic use , Sex Factors , Somatostatin/blood , Somatostatin/therapeutic use
19.
Cancer Chemother Pharmacol ; 53(5): 433-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15132131

ABSTRACT

PURPOSE: Imatinib (Glivec) has been established as a highly effective therapy for chronic myeloid leukemia and gastrointestinal tumors. The recommended daily dosage of 400-600 mg requires simultaneous intake of up to six of the current 100-mg capsules. Due to the need to swallow multiple capsules per dose, there is a potential negative impact on treatment adherence; therefore, a new imatinib 400-mg film-coated tablet has been developed. To improve dosing flexibility, particularly with regard to the pediatric population and the management of adverse events, a scored 100-mg film-coated tablet has also been introduced. EXPERIMENTAL DESIGN: A group of 33 healthy subjects were randomly assigned to one of six treatment sequences, in which they received imatinib as 4 x 100-mg capsules (reference), 4 x 100-mg scored tablets (test), and 1 x 400-mg tablet (test). Blood sampling was performed for up to 96 h after dosing, followed by a 10-day washout period prior to the next sequence. After the third dosing, subjects were monitored to assess delayed drug-related adverse events. Pharmacokinetic parameters were assessed using concentration-time curves for plasma imatinib and its metabolite CGP74588. RESULTS: Median Tmax was 2.5 h for capsules and tablets. Mean AUC((0-inf)) values were 27,094, 26,081 and 25,464 ng.h/ml for 4 x 100-mg capsules, 4 x 100-mg tablets, and 1 x 400-mg tablets, respectively. Cmax values were 1748, 1638 and 1606 ng/ml, and t(1/2) values were 15.8, 15.9 and 15.7 h. The test/reference ratios for AUC((0-inf)), AUC((0-96) (h)), and C(max) were 0.98, 0.98 and 0.95 for 4 x 100-mg tablets versus 4 x 100-mg capsules, and 0.95, 0.95 and 0.92 for 1 x 400-mg tablet versus 4 x 100-mg capsules. The 95% confidence intervals were fully contained within the interval (0.80, 1.25). Eight mild and one moderate adverse event considered to be drug related were reported. These events showed no clustering by type of dosage form and were of little to no clinical significance. CONCLUSIONS: Film-coated 100-mg (scored) and 400-mg tablet dose forms of imatinib are bioequivalent to the commercial 100-mg hard-gelatin capsule, and are as safe and well tolerated.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Piperazines/pharmacokinetics , Pyrimidines/pharmacokinetics , Adolescent , Adult , Aged , Benzamides , Cross-Over Studies , Female , Humans , Imatinib Mesylate , Male , Middle Aged , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Tablets , Therapeutic Equivalency
20.
J Clin Pharmacol ; 43(2): 154-62, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12616668

ABSTRACT

An open-label pharmacokinetic and pharmacodynamic study of zoledronic acid (Zometa) was performed in 19 cancer patients with bone metastases and known, varying levels of renal function. Patients were stratified according to creatinine clearance (CLcr) into different groups of normal (CLcr > 80 mL/min), mildly (CLcr = 50-80 mL/min), or moderately/severely impaired (CLcr = 10-50 mL/min) renal function. Three intravenous infusions of 4 mg zoledronic acid were administered at 1-month intervals between doses. Plasma concentrations and amounts excreted in urine were determined in all subjects, and 4 patients were administered 14C-labeled zoledronic acid to assess excretion and distribution of drug in whole blood. In general, the drug was well tolerated by the patients. Mean area under the plasma concentration versus time curve and mean concentration immediately after cessation of drug infusion were lower, and mean amounts excreted in urine over 24 hours from start of infusion were higher in normal subjects than in those with impaired renal function (36% vs. 28% of excreted dose), although the differences were not significant. Furthermore, with repeated doses, there was no evidence of drug accumulation in plasma or changes in drug exposure in any of the groups, nor was there any evidence of changes in renal function status. Serum levels of markers of bone resorption (serum C-telopeptide and N-telopeptide) were noticeably reduced after each dose of zoledronic acid across all three renal groups. It was concluded that in patients with mildly to moderately reduced renal function, dosage adjustment of zoledronic acid is likely not necessary.


Subject(s)
Bone Neoplasms/secondary , Diphosphonates , Imidazoles , Kidney/drug effects , Adult , Aged , Area Under Curve , Bone Neoplasms/metabolism , Diphosphonates/blood , Diphosphonates/pharmacokinetics , Diphosphonates/pharmacology , Female , Humans , Imidazoles/blood , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Infusions, Intravenous , Kidney/metabolism , Kidney Function Tests , Male , Metabolic Clearance Rate , Middle Aged , Zoledronic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...