Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Cancer ; 14(9): 1571-1578, 2023.
Article in English | MEDLINE | ID: mdl-37325049

ABSTRACT

The pharmacokinetics (PK) and safety of single-dose alpelisib (300 mg) were assessed in participants with moderate to severe hepatic impairment (n = 6 each) compared with their matching healthy controls (n = 11). Blood samples were collected upto 144 hours post-dose and evaluated by liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay. The primary PK parameters (maximum plasma concentration [Cmax], area under the curve [AUC]inf and AUClast) and secondary PK parameters (AUC0-t, apparent total body clearance [CL/F], apparent volume of distribution [Vz/F], time of maximum observed concentration [Tmax], and half-life [T1/2]) of oral alpelisib 300 mg were determined from individual plasma concentration-time profiles using non­compartmental analysis. Cmax of alpelisib decreased by approximately 17% in the moderate hepatic impairment group vs. the healthy control group (geometric mean ratio; GMR [90% confidence interval; CI], 0.833 [0.530, 1.31]). Cmax in the severe hepatic impairment group was comparable to that of the healthy control group (GMR [90% CI], 1.00 [0.636, 1.58]). AUClast for alpelisib decreased by approximately 27% in the moderate hepatic impairment group vs. the healthy control group (GMR [90% CI], 0.726 [0.487, 1.08]). AUClast was 26% higher in the severe hepatic impairment group compared with the healthy control group (GMR [90% CI], 1.26 [0.845, 1.87]). Overall, 3 participants (13.0%) experienced at least 1 adverse event which were either grade 1 or 2. Adverse events did not lead to study drug discontinuation. No grade 3 or 4 adverse events, serious adverse events or deaths were reported. The results indicate that a single dose of alpelisib was well tolerated in this study population. There was no significant impact of moderate or severe hepatic impairment on the exposure of alpelisib.

3.
Clin Pharmacol Ther ; 112(5): 1040-1050, 2022 11.
Article in English | MEDLINE | ID: mdl-35776072

ABSTRACT

Asciminib (Scemblix) is a first-in-class BCR::ABL1 inhibitor that works by specifically targeting the ABL myristoyl pocket (STAMP) and has potent activity against the T315I mutation. This study aimed to characterize the effect of asciminib exposure on disease progression and to elucidate factors influencing efficacy. Our analysis included 303 patients with chronic myeloid leukemia in chronic phase recruited in a phase I study with dose ranging from 10 to 200 mg twice a day (b.i.d.) or 40 to 200 mg once a day (q.d.) (NCT02081378) and in the phase III ASCEMBL (Study of Efficacy of CML-CP Patients Treated With ABL001 Versus Bosutinib, Previously Treated With 2 or More TKIs) study receiving asciminib 40 mg b.i.d. (NCT03106779). A total of 67 patients harbored the T315I mutation. A longitudinal pharmacokinetic/pharmacodynamic model was developed to characterize the exposure-efficacy relationship, in which the efficacy was assessed through BCR::ABL1 transcript levels over time. Specifically, a three-compartment model representing quiescent leukemic stem cells, proliferating bone marrow cells, and resistant cells was developed. Drug killing of the proliferating cells by asciminib was characterized by a power model. A subgroup analysis was performed on the patients with the T315I mutation using a maximum drug effect model to characterize the drug effect. The model demonstrated the appropriateness of a total daily dose of asciminib 80 mg in patients without the T315I mutation and 200 mg b.i.d. in patients with the T315I mutation with further validation in light of safety data. This model captured key characteristics of patients' response to asciminib and helped inform dosing rationale for resistant and difficult-to-treat populations.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Philadelphia Chromosome , Humans , Drug Resistance, Neoplasm/genetics , Mutation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/pharmacology
4.
Clin Pharmacokinet ; 61(10): 1393-1403, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35764773

ABSTRACT

BACKGROUND: Asciminib, a first-in-class, highly potent and specific ABL/BCR-ABL1 inhibitor, has shown superior efficacy compared to bosutinib in patients with Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase, treated with two or more tyrosine kinase inhibitors. This study aimed to describe pharmacokinetic (PK) properties of asciminib and to identify clinically relevant covariates impacting its exposure. METHODS: A population PK (PopPK) model was developed using a two-compartment model with delayed first-order absorption and elimination. The analysis included PK data from two clinical studies (Phases 1 and 3) involving 353 patients, with total daily dose of asciminib in the range of 20-400 mg. RESULTS: The nominal total daily dose was incorporated as a structural covariate on clearance (CL), and body weight (BW) was included as a structural covariate via allometric scaling on CL and central volume. Renal function and formulation were included as statistically significant covariates on CL and absorption (ka), respectively. The simulation results revealed a modest but clinically non-significant effect of baseline BW and renal function on ka. Correlations between covariates, such as baseline demographics and disease characteristics, heavy smoking status, hepatic function, and T315I mutation status, were not statistically significant with respect to CL, and they were not incorporated in the final model. Additionally, the final model-based simulations demonstrated comparable exposure and CL for asciminib 40 mg twice daily and 80 mg once daily (an alternative regimen not studied in the Phase 3 trial), as well as similar PK properties in patients with and without the T315I mutation. CONCLUSIONS: The final PopPK model adequately characterized the PK properties of asciminib and assessed the impact of key covariates on its exposure. The model corroborates the use of the approved asciminib dose of 80 mg total daily dose as 40 mg twice daily, and supports the use of 80 mg once daily as an alternative dose regimen to facilitate patient's compliance. TRIAL REGISTRATION NUMBER [DATE OF REGISTRATION]: First-in-human (CABL001X2101, Phase 1), ClinicalTrials.gov identifier: NCT02081378 [28 February 2014]; ASCEMBL (CABL001A2301, Phase 3), ClinicalTrials.gov identifier: NCT03106779 [10 April 2017].


Subject(s)
Antineoplastic Agents , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/therapeutic use , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Niacinamide/analogs & derivatives , Philadelphia Chromosome , Protein Kinase Inhibitors/therapeutic use , Pyrazoles
5.
AAPS J ; 22(6): 134, 2020 10 18.
Article in English | MEDLINE | ID: mdl-33070288

ABSTRACT

A physiologically based pharmacokinetic (PBPK) human model for alpelisib, an oral α-specific class I phosphatidylinositol-3-kinase (PI3K) inhibitor, was established to simulate oral absorption and plasma pharmacokinetics of healthy subjects to allow model-informed drug development. The GastroPlus™ model consisted of an advanced absorption gut model, which was linked to a 2-compartmental model. Systemic clearance and volume of distribution were estimated using population pharmacokinetics (popPK). Various food effect and pH-mediated absorption drug-drug interaction (DDI) scenarios were modeled. In fasted healthy subjects, simulated absorption was lower (ca. 70% for a 300-mg dose) due to pH and bile acid concentration-dependent solubility. Ranitidine showed a significant pH-mediated DDI effect only in the fasted but not fed state. The PBPK model identified that more drug is absorbed in the fed state, and alpelisib intestinal permeability is rate limiting to systemic exposure. Simulations for healthy subject showed a positive food effect with ca. 2-fold increase in plasma Cmax and 1.5-fold increase in AUC0-inf with a meal compared with fasted conditions. The PBPK model was verified using clinical food effect data with pivotal clinical formulation (PCF) and then applied to predict the performance of a commercial formulation (CF) in healthy volunteers. The model successfully predicted the outcome of a clinical bioequivalence study for PCF and CF with included in vitro dissolution data, both fasted and fed state. Estimated predictive errors (based on plasma Cmax, AUC0-t) were equal or below 30%. The alpelisib model for healthy subjects enables future bioequivalence formulation assessments, in fasted, fed, or altered pH conditions. Graphical Abstract.


Subject(s)
Food-Drug Interactions , Intestinal Absorption/physiology , Models, Biological , Phosphoinositide-3 Kinase Inhibitors/pharmacokinetics , Thiazoles/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Caco-2 Cells , Cross-Over Studies , Dogs , Drug Evaluation, Preclinical , Fasting/physiology , Female , Healthy Volunteers , Humans , Hydrogen-Ion Concentration , Male , Permeability , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Phosphoinositide-3 Kinase Inhibitors/adverse effects , Rats , Solubility , Tablets , Therapeutic Equivalency , Thiazoles/administration & dosage , Thiazoles/adverse effects , Young Adult
6.
J Clin Pharmacol ; 56(3): 316-23, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26183800

ABSTRACT

The pharmacokinetics (PK) and safety of single-dose buparlisib (30 mg) were assessed in subjects with mild to severe hepatic impairment (n = 6 each) relative to healthy controls (n = 13). Blood samples were collected until 336 hours postdose and evaluated by liquid chromatography tandem mass spectrometry. PK parameters (including area under the curve [AUC∞ ] and Cmax ) were derived using noncompartmental analysis. Buparlisib was rapidly absorbed in all groups (median Tmax 1.0-1.3 h). Buparlisib exposure (AUC∞ ) was moderately increased in subjects with mild (geometric mean ratio [GMR] 1.16; 90%CI 0.81, 1.65), moderate (GMR 1.14; 90%CI 0.80, 1.63), or severe (GMR 1.20; 90%CI 0.84, 1.72) hepatic impairment, relative to healthy controls. Apparent oral clearance was similar across groups. Due to a higher unbound fraction in the severe group (0.21) than all other groups (0.17), subjects with severe hepatic impairment had greater exposure to unbound buparlisib (GMR relative to healthy controls: AUC∞ 1.52; 90%CI 1.09, 2.13; Cmax 1.83; 90%CI 1.42, 2.36). The results indicate that a buparlisib dose adjustment may not be necessary for patients with mild to moderate hepatic impairment. The safety and therapeutic indices should be considered before determining if a dose adjustment is appropriate for patients with severe hepatic impairment.


Subject(s)
Aminopyridines/administration & dosage , Aminopyridines/pharmacokinetics , Liver Diseases/blood , Liver Diseases/diagnosis , Morpholines/administration & dosage , Morpholines/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Aminopyridines/adverse effects , Aminopyridines/blood , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Case-Control Studies , Female , Humans , Male , Middle Aged , Morpholines/adverse effects , Morpholines/blood , Severity of Illness Index , Young Adult
7.
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
8.
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
9.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...