Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
2.
Drug Metab Dispos ; 46(2): 109-121, 2018 02.
Article in English | MEDLINE | ID: mdl-29117990

ABSTRACT

Midostaurin (PKC412) is being investigated for the treatment of acute myeloid leukemia (AML) and advanced systemic mastocytosis (advSM). It is extensively metabolized by CYP3A4 to form two major active metabolites, CGP52421 and CGP62221. In vitro and clinical drug-drug interaction (DDI) studies indicated that midostaurin and its metabolites are substrates, reversible and time-dependent inhibitors, and inducers of CYP3A4. A simultaneous pharmacokinetic model of parent and active metabolites was initially developed by incorporating data from in vitro, preclinical, and clinical pharmacokinetic studies in healthy volunteers and in patients with AML or advSM. The model reasonably predicted changes in midostaurin exposure after single-dose administration with ketoconazole (a 5.8-fold predicted versus 6.1-fold observed increase) and rifampicin (90% predicted versus 94% observed reduction) as well as changes in midazolam exposure (1.0 predicted versus 1.2 observed ratio) after daily dosing of midostaurin for 4 days. The qualified model was then applied to predict the DDI effect with other CYP3A4 inhibitors or inducers and the DDI potential with midazolam under steady-state conditions. The simulated midazolam area under the curve ratio of 0.54 and an accompanying observed 1.9-fold increase in the CYP3A4 activity of biomarker 4ß-hydroxycholesterol indicated a weak-to-moderate CYP3A4 induction by midostaurin and its metabolites at steady state in patients with advSM. In conclusion, a simultaneous parent-and-active-metabolite modeling approach allowed predictions under steady-state conditions that were not possible to achieve in healthy subjects. Furthermore, endogenous biomarker data enabled evaluation of the net effect of midostaurin and its metabolites on CYP3A4 activity at steady state and increased confidence in DDI predictions.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Drug Interactions/physiology , Staurosporine/analogs & derivatives , Adult , Biomarkers/metabolism , Cytochrome P-450 CYP3A Inducers/metabolism , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Female , Humans , Hydroxycholesterols/metabolism , Ketoconazole/metabolism , Ketoconazole/pharmacokinetics , Male , Midazolam/metabolism , Midazolam/pharmacokinetics , Middle Aged , Models, Biological , Rifampin/metabolism , Rifampin/pharmacokinetics , Staurosporine/metabolism , Staurosporine/pharmacokinetics , Young Adult
3.
Clin Pharmacol Drug Dev ; 6(1): 19-26, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27138546

ABSTRACT

The objectives of this analysis were to establish the exposure-response relationship between plasma rifampicin and 4ß-hydroxycholesterol (4ßHC) concentration and to estimate the effect of weak, moderate, and potent CYP3A induction. Plasma rifampicin and 4ßHC concentration-time data from a drug-drug interaction study with rifampicin 600 mg were used for model development. An indirect response model with an effect compartment described the relationship between rifampicin and 4ßHC concentrations. The model predicted that the equilibration t1/2 and 4ßHC t1/2 were 72.8 and 142 hours, respectively. EM50 and Emax of rifampicin induction were 32.6 µg and 8.39-fold, respectively. The population PK-PD model was then used to simulate the effects of rifampicin 10, 20, and 100 mg on plasma 4ßHC for up to 21 days, in which rifampicin 10, 20, and 100 mg were used to represent weak, moderate, and strong inducers, respectively. The model-predicted median (5th, 95th percentiles) 1.13 (1.04, 1.44)-, 1.28 (1.10, 1.71)-, and 2.10 (1.45, 3.49)-fold increases in plasma 4ßHC after 14-day treatment with rifampicin 10, 20, and 100 mg, respectively. A new drug candidate can likely be classified as a weak, moderate, or strong inducer if baseline-normalized plasma 4ßHC increases by <1.13-, 1.13- to 2.10-, or >2.10-fold, respectively, after 14 days of dosing.


Subject(s)
Cytochrome P-450 CYP3A Inducers/administration & dosage , Hydroxycholesterols/blood , Rifampin/administration & dosage , Adult , Cytochrome P-450 CYP3A Inducers/blood , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Drug Interactions , Female , Healthy Volunteers , Humans , Hydroxycholesterols/administration & dosage , Hydroxycholesterols/pharmacology , Male , Middle Aged , Models, Theoretical , Rifampin/blood , Rifampin/pharmacokinetics , Young Adult
4.
Eur J Clin Pharmacol ; 70(8): 915-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24839948

ABSTRACT

PURPOSE: Midostaurin, a multitargeted tyrosine kinase inhibitor, is primarily metabolized by CYP3A4. This midostaurin drug-drug interaction study assessed the dynamic response and clinical usefulness of urinary 6ß-hydroxycortisol to cortisol ratio (6ßCR) and plasma 4ß-hydroxycholesterol (4ßHC) for monitoring CYP3A4 activity in the presence or absence of rifampicin, a strong CYP3A4 inducer. METHODS: Forty healthy adults were randomized into groups for either placebo or treatment with rifampicin 600 mg QD for 14 days. All participants received midostaurin 50 mg on day 9. Midostaurin plasma pharmacokinetic parameters were assessed. Urinary 6ßCR and plasma 4ßHC levels were measured on days 1, 9, 11, and 15. RESULTS: Both markers remained stable over time in the control group and increased significantly in the rifampicin group. In the rifampicin group, the median increases (vs day 1) on days 9, 11, and 15 were 4.1-, 5.2-, and 4.7-fold, respectively, for 6ßCR and 3.4-, 4.1-, and 4.7-fold, respectively, for 4ßHC. Inter- and intrasubject variabilities in the control group were 45.6 % and 30.5 %, respectively, for 6ßCR, and 33.8 % and 7.5 %, respectively, for 4ßHC. Baseline midostaurin area under the concentration-time curve (AUC) correlated with 4ßHC levels (ρ = -0.72; P = .003), but not with 6ßCR (ρ = 0.0925; P = .6981). CONCLUSIONS: Both 6ßCR and 4ßHC levels showed a good dynamic response range upon strong CYP3A4 induction with rifampicin. Because of lower inter- and intrasubject variability, 4ßHC appeared more reliable and better predictive of CYP3A4 activity compared with 6ßCR. The data from our study further support the clinical utility of these biomarkers.


Subject(s)
Cytochrome P-450 CYP3A Inducers/pharmacology , Hydrocortisone/analogs & derivatives , Hydroxycholesterols/blood , Protein Kinase Inhibitors/pharmacokinetics , Rifampin/pharmacology , Staurosporine/analogs & derivatives , Adult , Biomarkers/blood , Biomarkers/urine , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Female , Humans , Hydrocortisone/urine , Male , Middle Aged , Protein Kinase Inhibitors/blood , Staurosporine/blood , Staurosporine/pharmacokinetics , Young Adult
5.
Cancer Chemother Pharmacol ; 72(6): 1223-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24085261

ABSTRACT

PURPOSE: Midostaurin (PKC412), a multitargeted tyrosine kinase inhibitor that targets FMS-related tyrosine kinase 3 and KIT, is in clinical trials for the treatment for acute myeloid leukemia and advanced systemic mastocytosis. In vitro studies showed that midostaurin is predominantly metabolized by cytochrome P450 3A4 (CYP3A4) and that midostaurin inhibits and/or induces the same enzyme. Here, we address the clinical relevance of CYP3A4-related drug-drug interactions with midostaurin as either a "victim" or "perpetrator." METHODS: Three phase I studies in healthy volunteers evaluated the effects of a CYP3A4 inhibitor (ketoconazole 400 mg daily for 10 days) or CYP3A4 inducer (rifampicin 600 mg daily for 14 days) on concentrations of midostaurin and its metabolites following a single 50-mg dose of midostaurin and the effects of midostaurin as a single dose (100 mg) and multiple doses (50 mg twice daily) on midazolam (a sensitive CYP3A4 probe) concentration. The plasma concentrations of midostaurin and its 2 active metabolites, CGP62221 and CGP52421, were determined using a sensitive liquid chromatography/tandem mass spectrometry method. RESULTS: Inhibition of CYP3A4 by ketoconazole increased midostaurin exposure more than tenfold, and induction of CYP3A4 by rifampicin decreased midostaurin exposure by more than tenfold. Midostaurin did not appreciably affect the concentrations of midazolam or its metabolite, 1'-hydroxymidazolam, at single or multiple doses. CONCLUSION: The pharmacokinetics of midostaurin and its metabolites was affected substantially by ketoconazole and rifampicin, suggesting that midostaurin is a sensitive CYP3A4 substrate. Midostaurin did not appear to inhibit or induce CYP3A4 in vivo.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Enzyme Inhibitors/pharmacology , Staurosporine/analogs & derivatives , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Chromatography, Liquid , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 CYP3A Inhibitors , Drug Administration Schedule , Drug Interactions , Enzyme Induction/drug effects , Enzyme Inhibitors/administration & dosage , Female , Humans , Ketoconazole/pharmacology , Male , Midazolam/pharmacology , Middle Aged , Rifampin/pharmacology , Staurosporine/administration & dosage , Staurosporine/pharmacokinetics , Staurosporine/pharmacology , Tandem Mass Spectrometry , Young Adult
6.
Pharmacotherapy ; 33(12): 1341-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23798029

ABSTRACT

PURPOSE: To determine the preclinical activity, clinical maximum tolerated dose (MTD), and recommended phase II dose of midostaurin (MS) combined either sequentially or concurrently with intravenous decitabine (DAC) in newly diagnosed patients 60 years or older or relapsed/refractory adult patients (18 years or older) with acute myeloid leukemia (AML). PATIENTS AND METHODS: Cultured and primary AML cells were treated with DAC and/or MS and analyzed by flow cytometry and immunoblot analyses. In the phase I study, 16 patients were enrolled; 8 were newly diagnosed patients 60 years or older and 8 were 18 years or older with relapsed AML. Only 2 of 16 patients (13%) had FLT3-internal tandem duplication (ITD) mutations, and no patient had KIT mutations. RESULTS: Compared with treatment with either agent alone, sequential treatment with DAC and MS exerted superior anti-AML activity in cultured and primary FLT3-ITD-expressing AML cells. In the subsequent phase I study, the MTD and schedule of administration of the combination was identified as DAC followed by MS. Three patients developed dose-limiting toxicities: two patients developed pulmonary edema requiring mechanical ventilation and one patient developed a prolonged QTc greater than 500 msec. Based on an intent-to-treat analysis, 57% of the patients achieved stable disease or better while enrolled in the trial; 25% had a complete hematologic response. Pharmacokinetic analysis revealed results similar to those previously reported for MS. CONCLUSION: The in vitro combination of DAC and MS is synergistically active against FLT3-ITD mutations expressing AML cells. In a clinical setting, the combination of sequentially administered DAC followed by MS is possible without significant unexpected toxicity, but the concurrent administration of DAC and MS led to pulmonary toxicity after only a few doses. On the basis of these results, additional studies exploring the sequential combination of untreated AML in elderly patients are warranted to further evaluate this combination at the MTD.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , fms-Like Tyrosine Kinase 3/genetics , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Cell Line, Tumor , Cohort Studies , Decitabine , Drug Synergism , Female , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Maximum Tolerated Dose , Middle Aged , Mutation , Recurrence , Staurosporine/administration & dosage , Staurosporine/analogs & derivatives
7.
Cancer Chemother Pharmacol ; 69(5): 1255-63, 2012 May.
Article in English | MEDLINE | ID: mdl-22294470

ABSTRACT

PURPOSE: Midostaurin (PKC412) is a multitargeted tyrosine kinase inhibitor of FMS-like tyrosine kinase 3 receptor (FLT3), c-KIT, and other receptors. Midostaurin is active in patients with acute myeloid leukemia and systemic mastocytosis. Although no substantive risk for cardiac abnormalities has been observed with midostaurin in clinical studies thus far, some TKIs have been shown to affect cardiac repolarization. Here we evaluated midostaurin's effect on cardiac repolarization. METHODS: This phase I study evaluated the effect of midostaurin (75 mg twice daily for 2 days; 75 mg once on day 3) on the heart rate-corrected QT (QTc) interval in a parallel design with active (moxifloxacin) and placebo control arms in healthy volunteers. RESULTS: The maximum mean QTc change from baseline corrected using Fridericia's correction (QTcF) for midostaurin compared with placebo was 0.7 ms at 24 h post dose on day 3. The highest upper bound of the 1-sided 95% CI was 4.7 ms, which excluded 10 ms, demonstrating a lack of QTcF prolongation effect. Assay sensitivity was demonstrated by modeling the moxifloxacin plasma concentration versus QTcF change from baseline, which showed a clear positive increase in QTcF with increasing moxifloxacin plasma concentrations, as expected based on previous studies. In the 4-day evaluation period, a minority of participants (34.6%) experienced an adverse event; 97.0% were grade 1. No grade 3 or 4 adverse events were reported. CONCLUSION: Midostaurin demonstrated a good safety profile in healthy volunteers, with no prolonged cardiac repolarization or other changes on the electrocardiogram.


Subject(s)
Antineoplastic Agents/adverse effects , Electrocardiography , Protein Kinase Inhibitors/adverse effects , Staurosporine/analogs & derivatives , Adolescent , Adult , Antineoplastic Agents/administration & dosage , Aza Compounds/adverse effects , Double-Blind Method , Female , Fluoroquinolones , Heart Rate , Humans , Long QT Syndrome/chemically induced , Male , Middle Aged , Moxifloxacin , Protein Kinase Inhibitors/administration & dosage , Quinolines/adverse effects , Staurosporine/administration & dosage , Staurosporine/adverse effects , Young Adult
8.
J Clin Oncol ; 28(28): 4339-45, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20733134

ABSTRACT

PURPOSE: Mutations leading to constitutive activation of the FMS-like tyrosine kinase 3 receptor (FLT3) occur in blasts of 30% of patients with acute myeloid leukemia (AML). Midostaurin (PKC412; N-benzoylstaurosporin) is a multitargeted tyrosine kinase inhibitor, with demonstrated activity in patients with AML/myelodysplastic syndrome (MDS) with FLT3 mutations. PATIENTS AND METHODS: Ninety-five patients with AML or MDS with either wild-type (n = 60) or mutated (n = 35) FLT3 were randomly assigned to receive oral midostaurin at 50 or 100 mg twice daily. The drug was discontinued in the absence of response at 2 months, disease progression, or unacceptable toxicity. Response was defined as complete response, partial response (PR), hematologic improvement, or reduction in peripheral blood or bone marrow blasts by ≥ 50% (BR). RESULTS: The rate of BR for the population in whom efficacy could be assessed (n = 92) was 71% in patients with FLT3-mutant and 42% in patients with FLT3 wild-type. One PR occurred in a patient with FLT3-mutant receiving the 100-mg dose regimen. Both doses were well-tolerated; there were no differences in toxicity or response rate according to the dose of midostaurin. CONCLUSION: These results suggest that midostaurin has hematologic activity in both patients with FLT3-mutant and wild-type. The degree of clinical activity observed supports additional studies that combine midostaurin and other agents such as chemotherapy especially in FLT3-mutant AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Staurosporine/analogs & derivatives , fms-Like Tyrosine Kinase 3/genetics , Administration, Oral , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Female , Humans , Leukemia, Myeloid, Acute/enzymology , Male , Mutation , Myelodysplastic Syndromes/enzymology , Receptor Protein-Tyrosine Kinases/genetics , Staurosporine/administration & dosage , Staurosporine/adverse effects , Staurosporine/therapeutic use , Treatment Outcome
9.
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
10.
J Clin Pharmacol ; 48(4): 428-35, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18281442

ABSTRACT

Deferasirox (ICL670) is representative of a new class of tridentate iron chelators, formulated as tablets for dispersion. Deferasirox has exhibited high potency and a clinically manageable safety profile in preclinical models and in an extensive clinical program. The effect of food and time of food intake on the pharmacokinetics of deferasirox was investigated in healthy volunteers and patients with transfusional hemosiderosis. The bioequivalence of a single oral dose of deferasirox (20 mg/kg) was assessed following administration either before a high-fat or standard breakfast or concurrent with a standard breakfast in comparison with fasted conditions in healthy volunteers. The bioavailability of deferasirox was determined following a single oral dose (20 mg/kg) under fed and fasted conditions in patients. These data show that the type of food, caloric content, and fat content of the meal influence the bioavailability of deferasirox when consumed concomitantly. In contrast, this is not the case when deferasirox is administered at least 30 minutes before a meal. In conclusion, it is recommended that deferasirox be administered at least 30 minutes prior to meals. When this is not feasible, deferasirox should be administered consistently at the same time before meals to limit the sources of variability that affect absorption.


Subject(s)
Benzoates/pharmacokinetics , Eating/physiology , Iron Chelating Agents/pharmacokinetics , Iron Overload/metabolism , Triazoles/pharmacokinetics , Adolescent , Adult , Area Under Curve , Benzoates/administration & dosage , Biological Availability , Chromatography, High Pressure Liquid , Cross-Over Studies , Deferasirox , Female , Food , Food-Drug Interactions , Humans , Intestinal Absorption , Iron Chelating Agents/administration & dosage , Male , Middle Aged , Spectrophotometry, Ultraviolet , Triazoles/administration & dosage
11.
Clin Cancer Res ; 13(14): 4164-9, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17634544

ABSTRACT

PURPOSE: To investigate the potential use of accelerator mass spectrometry (AMS) in the study of the clinical pharmacology of imatinib. EXPERIMENTAL DESIGN: Six patients who were receiving imatinib (400 mg/d) as part of their ongoing treatment for chronic myeloid leukemia (CML) received a dose containing a trace quantity (13.6 kBq) of (14)C-imatinib. Blood samples were collected from patients before and at various times up to 72 h after administration of the test dose and were processed to provide samples of plasma and peripheral blood lymphocytes (PBL). Samples were analyzed by AMS, with chromatographic separation of parent compound from metabolites. In addition, plasma samples were analyzed by liquid chromatography/mass spectrometry (LCMS). RESULTS: Analysis of the AMS data indicated that imatinib was rapidly absorbed and could be detected in plasma up to 72 h after administration. Imatinib was also detectable in PBL at 24 h after administration of the (14)C-labeled dose. Comparison of plasma concentrations determined by AMS with those derived by LCMS analysis gave similar average estimates of area under plasma concentration time curve (26 +/- 3 versus 27 +/- 11 microg/mL.h), but with some variation within each individual. CONCLUSIONS: Using this technique, data were obtained in a small number of patients on the pharmacokinetics of a single dose of imatinib in the context of chronic dosing, which could shed light on possible pharmacologic causes of resistance to imatinib in CML.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Adult , Benzamides , Female , Humans , Imatinib Mesylate , Male , Mass Spectrometry , Middle Aged , Particle Accelerators , Polymerase Chain Reaction
12.
Anticancer Drugs ; 18(2): 219-25, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17159608

ABSTRACT

LAF389 is a synthetic analogue of bengamide B, a natural product isolated from Jaspidae sponges. LAF389 has both antiproliferative and antiangiogenetic properties, and preclinical investigations showed a broad antitumour activity. This clinical trial aimed to determine the safety and pharmacokinetic profile of LAF389 administered as a slow intravenous injection for 3 consecutive days every 3 weeks in patients with advanced solid tumours. Eight dose levels were tested: 1, 2.5, 5, 10, 15, 30, 25 and 20 mg/day. A total of 33 patients, median age 52 years (range 33-72), with refractory solid tumours were enroled, 19 men and 14 women with a median World Health Organization performance status of 1 (0-4). Seventy-eight cycles of treatment have been administered (mean 2.5, range 1-10). Four cardiovascular dose-limiting toxicities were reported at 30 mg (2/2 patients) and 25 mg (2/9 patients), eight additional patients at various dose levels had (cardio)vascular toxicity, probably drug related, and one patient died owing to pulmonary embolism at the 5 mg dose. No objective responses were recorded. Pharmacokinetic parameters were variable, although linear and without obvious accumulation from cycle I to cycle II. LAF389 dose escalation was terminated owing to occurrence of unpredictable cardiovascular events. This, associated with the lack of clinical activity, did not warrant further investigation of this agent.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/therapeutic use , Azepines/pharmacokinetics , Azepines/therapeutic use , Adult , Aged , Angiogenesis Inhibitors/adverse effects , Area Under Curve , Azepines/adverse effects , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/epidemiology , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/drug therapy
13.
Cancer Chemother Pharmacol ; 54(4): 290-4, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15138710

ABSTRACT

The study under discussion was a drug-drug interaction study in which the effect of ketoconazole, a potent CYP450 3A4 inhibitor, on the pharmacokinetics of Glivec (imatinib) was investigated. A total of 14 healthy subjects (13 male, 1 female) were enrolled in this study. Each subject received a single oral dose of imatinib 200 mg alone, and a single oral dose of imatinib 200 mg coadministered with a single oral dose of ketoconazole 400 mg according to a two-period crossover design. The treatment sequence was randomly allocated. Subtherapeutic imatinib doses and a short exposure were tested in order not to overexpose the healthy volunteers. There was a minimum 7-day washout period between the two sequences. Blood samples for determination of plasma concentrations were taken up to 96 h after dosing. Imatinib and CGP74588 (main metabolite of imatinib) concentrations were measured using LC/MS/MS method and pharmacokinetic parameters were estimated by a non-compartmental analysis. Following ketoconazole coadministration, the mean imatinib C(max), AUC((0-24)) and AUC((0- infinity )) increased significantly by 26% ( P<0.005), 40% ( P<0.0005) and 40% ( P <0.0005), respectively. There was a statistically significant decrease in apparent clearance (CL/f) of imatinib with a mean reduction of 28.6% ( P<0.0005). The mean C(max) and AUC((0-24)) of the metabolite CGP74588 decreased significantly by 22.6% ( P<0.005) and 13% ( P<0.05) after ketoconazole treatment, although the AUC((0- infinity )) of CGP74588 only decreased by 5% ( P=0.28). Coadministration of ketoconazole and imatinib caused a 40% increase in exposure to imatinib in healthy volunteers. Given its previously demonstrated safety profile, this increased exposure to imatinib is likely to be clinically significant only at high doses. This interaction should be considered when administering inhibitors of the CYP3A family in combination with imatinib.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Ketoconazole/pharmacokinetics , Piperazines/pharmacokinetics , Pyrimidines/pharmacokinetics , Adult , Benzamides , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Drug Interactions , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Imatinib Mesylate , Ketoconazole/adverse effects , Male , Middle Aged , Piperazines/adverse effects , Pyrimidines/adverse effects
14.
J Clin Pharmacol ; 44(2): 158-62, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14747424

ABSTRACT

The purpose of this study was to investigate the absolute bioavailability of a single oral dose of imatinib (Glivec), 400 mg (capsules vs. oral solution), compared with imatinib, 100 mg (intravenous [i.v.] infusion), in healthy subjects. Twelve subjects received a single treatment in each treatment period: a 400-mg oral dose of imatinib in capsule form or as a solution or a 100-mg i.v. infusion of imatinib. Plasma imatinib concentrations were measured following each treatment; pharmacokinetic parameters and absolute bioavailability were determined. Absolute bioavailability values (compared with i.v. infusion) for the imatinib capsule and oral solution were 98.3% and 97.2%, respectively. Both the rate and extent of imatinib absorption, as measured by C(max), partial AUC, and total AUC, were similar for the oral solution and the imatinib capsule intended for the market. The 400-mg oral dose of imatinib, as a capsule or a solution, was completely absorbed and was almost completely bioavailable (> 97%).


Subject(s)
Enzyme Inhibitors/pharmacokinetics , Piperazines/pharmacokinetics , Pyrimidines/pharmacokinetics , Adult , Area Under Curve , Benzamides , Biological Availability , Capsules , Cross-Over Studies , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/blood , Female , Half-Life , Humans , Imatinib Mesylate , Infusions, Intravenous , Intestinal Absorption , Male , Metabolic Clearance Rate , Middle Aged , Pilot Projects , Piperazines/administration & dosage , Piperazines/blood , Pyrimidines/administration & dosage , Pyrimidines/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...