Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Br J Clin Pharmacol ; 84(5): 876-887, 2018 05.
Article in English | MEDLINE | ID: mdl-29346838

ABSTRACT

AIMS: The aims of the present study were to assess the safety, pharmacokinetics (PK) and pharmacodynamics (PD) of BMS-962212, a first-in-class factor XIa inhibitor, in Japanese and non-Japanese healthy subjects. METHODS: This was a randomized, placebo-controlled, double-blind, sequential, ascending-dose study of 2-h (part A) and 5-day (part B) intravenous (IV) infusions of BMS-962212. Part A used four doses (1.5, 4, 10 and 25 mg h-1 ) of BMS-962212 or placebo in a 6:2 ratio per dose. Part B used four doses (1, 3, 9 and 20 mg h-1 ) enrolling Japanese (n = 4 active, n = 1 placebo) and non-Japanese (n = 4 active, n = 1 placebo) subjects per dose. The PK, PD, safety and tolerability were assessed throughout the study. RESULTS: BMS-962212 was well tolerated; there were no signs of bleeding, and adverse events were mild. In parts A and B, BMS-962212 demonstrated dose proportionality. The mean half-life in parts A and B ranged from 2.04 to 4.94 h and 6.22 to 8.65 h, respectively. Exposure-dependent changes were observed in the PD parameters, activated partial thromboplastin time (aPTT) and factor XI clotting activity (FXI:C). The maximum mean aPTT and FXI:C change from baseline at 20 mg h-1 in part B was 92% and 90%, respectively. No difference was observed in weight-corrected steady-state concentrations, aPTT or FXI:C between Japanese and non-Japanese subjects (P > 0.05). CONCLUSION: BMS-962212 has tolerability, PK and PD properties suitable for investigational use as an acute antithrombotic agent in Japanese or non-Japanese subjects.


Subject(s)
Isoquinolines/adverse effects , Isoquinolines/pharmacology , Isoquinolines/pharmacokinetics , para-Aminobenzoates/adverse effects , para-Aminobenzoates/pharmacology , para-Aminobenzoates/pharmacokinetics , Adolescent , Adult , Asian People/statistics & numerical data , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fibrinolytic Agents/pharmacology , Healthy Volunteers , Humans , Infusions, Intravenous , Isoquinolines/administration & dosage , Male , Middle Aged , Partial Thromboplastin Time/statistics & numerical data , White People/statistics & numerical data , Young Adult , para-Aminobenzoates/administration & dosage
2.
Br J Clin Pharmacol ; 79(5): 838-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25377242

ABSTRACT

AIM: Apixaban is an orally active inhibitor of coagulation factor Xa and is eliminated by multiple pathways, including renal and non-renal elimination. Non-renal elimination pathways consist of metabolism by cytochrome P450 (CYP) enzymes, primarily CYP3A4, as well as direct intestinal excretion. Two single sequence studies evaluated the effect of ketoconazole (a strong dual inhibitor of CYP3A4 and P-glycoprotein [P-gp]) and diltiazem (a moderate CYP3A4 inhibitor and a P-gp inhibitor) on apixaban pharmacokinetics in healthy subjects. METHOD: In the ketoconazole study, 18 subjects received apixaban 10 mg on days 1 and 7, and ketoconazole 400 mg once daily on days 4-9. In the diltiazem study, 18 subjects received apixaban 10 mg on days 1 and 11 and diltiazem 360 mg once daily on days 4-13. RESULTS: Apixaban maximum plasma concentration and area under the plasma concentration-time curve extrapolated to infinity increased by 62% (90% confidence interval [CI], 47, 78%) and 99% (90% CI, 81, 118%), respectively, with co-administration of ketoconazole, and by 31% (90% CI, 16, 49%) and 40% (90% CI, 23, 59%), respectively, with diltiazem. CONCLUSION: A 2-fold and 1.4-fold increase in apixaban exposure was observed with co-administration of ketoconazole and diltiazem, respectively.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacology , Diltiazem/pharmacology , Factor Xa Inhibitors/pharmacokinetics , Ketoconazole/pharmacology , Pyrazoles/pharmacokinetics , Pyridones/pharmacokinetics , Adolescent , Adult , Cross-Over Studies , Cytochrome P-450 CYP3A/metabolism , Diltiazem/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/blood , Factor Xa Inhibitors/pharmacology , Female , Healthy Volunteers , Humans , Ketoconazole/administration & dosage , Male , Middle Aged , Pyrazoles/administration & dosage , Pyrazoles/blood , Pyrazoles/pharmacology , Pyridones/administration & dosage , Pyridones/blood , Pyridones/pharmacology , Young Adult
3.
Br J Clin Pharmacol ; 78(5): 1122-34, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24837659

ABSTRACT

AIMS: This study aimed to assess changes in the plasma concentrationss of 4ß-hydroxycholesterol (4ßHC) against intravenous (i.v.) and oral midazolam (MDZ) pharmacokinetics (PK) after administration of a potent CYP3A inhibitor [ketoconazole (KETO)] and inducer [rifampicin (RIF)]. METHODS: Thirty-two healthy subjects (HS) were allocated into three groups of 12 each in KETO and RIF and 10 in a placebo group (PLB). All HS were randomized to receive oral and i.v. MDZ on day 1 or 2 and on day 15 or 16 after receiving RIF (600 mg once daily), KETO (400 mg once daily) or PLB for 2 weeks. Subjects were followed until day 30. The effect of treatments on 4ßHC was assessed by analyzing % change from baseline using a linear spline mixed effects model. RESULTS: Compared with PLB, KETO decreased 4ßHC mean values up to 13% (P = 0.003) and RIF increased 4ßHC mean values up to 220% (P < 0.001). Within 14 days of stopping KETO and RIF, 4ßHC had either returned to baseline (KETO) or was still returning to baseline (RIF). Compared with baseline, mean oral MDZ AUC increased by 11-fold (90% CI ranging from 9-fold to 13-fold increase) and decreased by 92% (90% CI ranging from 90% to 95% decrease) after KETO and RIF, respectively. Similar trends were observed for 6ß-hydroxycortisol : cortisol (6ßHCL : CL) urinary ratios. CONCLUSIONS: Changes in plasma 4ßHC can be utilized as a surrogate for MDZ PK after multiple doses of potent CYP3A inducers. There is a more limited dynamic range for 4ßHC for assessment of potential CYP3A inhibitors. 4ßHC is a valuable tool for the assessment of potential CYP3A inducers in early drug development.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Hydroxycholesterols/blood , Midazolam/pharmacokinetics , Adolescent , Adult , Biomarkers/blood , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Injections, Intravenous , Ketoconazole/pharmacology , Limit of Detection , Midazolam/administration & dosage , Midazolam/blood , Middle Aged , Rifampin/pharmacology , Saliva/chemistry , Substrate Specificity , Time Factors , Tissue Distribution , Young Adult
4.
Br J Clin Pharmacol ; 78(4): 877-85, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24697979

ABSTRACT

AIM: To assess pharmacokinetic and pharmacodynamic interactions between naproxen (a non-steroidal anti-inflammatory drug) and apixaban (an oral, selective, direct factor-Xa inhibitor). METHOD: In this randomized, three period, two sequence study, 21 healthy subjects received a single oral dose of apixaban 10 mg, naproxen 500 mg or co-administration of both. Blood samples were collected for determination of apixaban and naproxen pharmacokinetics and pharmacodynamics (anti-Xa activity, international normalized ratio [INR] and arachidonic acid-induced platelet aggregation [AAI-PA]). Adverse events, bleeding time and routine safety assessments were also evaluated. RESULTS: Apixaban had no effect on naproxen pharmacokinetics. However, following co-administration, apixaban AUC(0,∞), AUC(0,t) and Cmax were 54% (geometric mean ratio 1.537; 90% confidence interval (CI) 1.394, 1.694), 55% (1.549; 90% CI 1.400, 1.713) and 61% (1.611; 90% CI 1.417, 1.831) higher, respectively. Mean (standard deviation [SD]) anti-Xa activity at 3 h post-dose was approximately 60% higher following co-administration compared with apixaban alone, 4.4 [1.0] vs. 2.7 [0.7] IU ml(-1) , consistent with the apixaban concentration increase following co-administration. INR was within the normal reference range after all treatments. AAI-PA was reduced by approximately 80% with naproxen. Co-administration had no impact beyond that of naproxen. Mean [SD] bleeding time was higher following co-administration (9.1 [4.1] min) compared with either agent alone (5.8 [2.3] and 6.9 [2.6] min for apixaban and naproxen, respectively). CONCLUSION: Co-administration of naproxen with apixaban results in higher apixaban exposure and appears to occur through increased apixaban bioavailability. The effects on anti-Xa activity, INR and inhibition of AAI-PA observed in this study were consistent with the individual pharmacologic effects of apixaban and naproxen.


Subject(s)
Factor Xa Inhibitors/pharmacokinetics , Naproxen/pharmacology , Pyrazoles/pharmacokinetics , Pyridones/pharmacokinetics , Adult , Drug Interactions , Female , Humans , International Normalized Ratio , Male , Pyrazoles/pharmacology , Pyridones/pharmacology
5.
Int J Clin Pharmacol Ther ; 52(7): 564-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24725442

ABSTRACT

OBJECTIVE: This was a randomized, placebo-controlled, double-blind, sequential, ascending-dose study to assess the safety, tolerability, pharmacokinetics, and pharmacodynamics of multiple oral doses of apixaban in healthy Japanese male subjects. METHODS: The study was conducted using three sequential dose panels: apixaban 2.5 mg, 5 mg, and 10 mg given twice daily. For each dose panel, subjects were randomly assigned to receive oral apixaban (n = 6) or matching placebo (n = 2) for 7 days. The pharmacokinetics of apixaban and effect on pharmacodynamic variables (clotting assays and anti-Xa activity) were assessed on day 1 and day 7 of treatment. Safety was assessed throughout the study. Only after the preceding dose was confirmed to be safe and well-tolerated subjects were enrolled into the next-higher-dose panel. RESULTS: Apixaban was safe and well-tolerated in these healthy Japanese male subjects across the doses evaluated. On day 7, peak plasma concentrations were reached ~ 3 hours postdose, and increases in peak plasma concentration (C(max)), trough plasma concentration, and area under the plasma concentration-time curve across one dosing interval (12 hours) were tested dose-proportional across the dose range. A modest degree of accumulation was observed that was similar for all doses (accumulation index of 1.7 to 2.0), and renal clearance was consistent across doses (0.91 L/h - 1.07 L/h). Exposure-dependent prolongation of prothrombin time, activated partial thromboplastin time, modified prothrombin time, and increases in anti-Xa activity were observed after single and multiple doses of apixaban. CONCLUSIONS: Apixaban was safe and well-tolerated in healthy Japanese subjects. The pharmacokinetic profile of apixaban following multiple twice-daily doses was linear, and exposure parameters such as C(max), observed at ~ 3 hours post-dose, and area under the plasma concentration-time curve increased in a dose-proportional manner. Pharmacodynamic profiles closely followed the apixaban plasma concentration-time profiles.


Subject(s)
Factor Xa Inhibitors , Pyrazoles/adverse effects , Pyridones/adverse effects , Adult , Double-Blind Method , Humans , Male , Prothrombin Time , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyridones/pharmacokinetics , Pyridones/pharmacology
6.
Br J Clin Pharmacol ; 76(6): 908-16, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23488672

ABSTRACT

AIM: Apixaban is an oral, direct, factor-Xa inhibitor approved for thromboprophylaxis in patients who have undergone elective hip or knee replacement surgery and for prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation. This open label, parallel group study investigated effects of extremes of body weight on apixaban pharmacokinetics, pharmacodynamics, safety and tolerability. METHOD: Fifty-four healthy subjects were enrolled [18 each into low (≤50 kg), reference (65-85 kg) and high (≥120 kg) body weight groups]. Following administration of a single oral dose of 10 mg apixaban, plasma and urine samples were collected for determination of apixaban pharmacokinetics and anti-factor Xa activity. Adverse events, vital signs and laboratory assessments were monitored. RESULTS: Compared with the reference body weight group, low body weight had approximately 27% [90% confidence interval (CI): 8-51%] and 20% (90% CI: 11-42%) higher apixaban maximum observed plasma concentration (Cmax) and area under the concentration-time curve extrapolated to infinity (AUC(0,∞)), respectively, and high body weight had approximately 31% (90% CI: 18-41%) and 23% (90% CI: 9-35%) lower apixaban Cmax and AUC(0,∞) , respectively. Apixaban renal clearance was similar across the weight groups. Plasma anti-factor Xa activity showed a direct, linear relationship with apixaban plasma concentration, regardless of body weight group. Apixaban was well tolerated in this study. CONCLUSION: The modest change in apixaban exposure is unlikely to require dose adjustment for apixaban based on body weight alone. However, caution is warranted in the presence of additional factors (such as severe renal impairment) that could increase apixaban exposure.


Subject(s)
Anticoagulants , Factor Xa Inhibitors , Ideal Body Weight , Overweight/metabolism , Pyrazoles , Pyridones , Thinness/metabolism , Administration, Oral , Adolescent , Adult , Anticoagulants/adverse effects , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Area Under Curve , Body Mass Index , Dose-Response Relationship, Drug , Female , Healthy Volunteers , Humans , Male , Middle Aged , Overweight/blood , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyridones/adverse effects , Pyridones/pharmacokinetics , Pyridones/pharmacology , Thinness/blood , Thrombosis/prevention & control , Young Adult
7.
Br J Clin Pharmacol ; 75(3): 763-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22823746

ABSTRACT

AIM: To determine the absolute oral bioavailability (F(p.o.) ) of saxagliptin and dapagliflozin using simultaneous intravenous ¹4C-microdose/therapeutic oral dosing (i.v.micro + oraltherap). METHODS: The F(p.o.) values of saxagliptin and dapagliflozin were determined in healthy subjects (n = 7 and 8, respectively) following the concomitant administration of single i.v. micro doses with unlabelled oraltherap doses. Accelerator mass spectrometry and liquid chromatography-tandem mass spectrometry were used to quantify the labelled and unlabelled drug, respectively. RESULTS: The geometric mean point estimates (90% confidence interval) F(p.o) . values for saxagliptin and dapagliflozin were 50% (48, 53%) and 78% (73, 83%), respectively. The i.v.micro had similar pharmacokinetics to oraltherap. CONCLUSIONS: Simultaneous i.v.micro + oraltherap dosing is a valuable tool to assess human absolute bioavailability.


Subject(s)
Adamantane/analogs & derivatives , Dipeptides/pharmacokinetics , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Glucosides/pharmacokinetics , Sodium-Glucose Transport Proteins/antagonists & inhibitors , Adamantane/pharmacokinetics , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Area Under Curve , Benzhydryl Compounds , Biological Availability , Chromatography, Liquid , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Dose-Response Relationship, Drug , Humans , Male , Mass Spectrometry , Middle Aged , Sodium-Glucose Transport Proteins/administration & dosage , Sodium-Glucose Transport Proteins/pharmacokinetics , White People , Young Adult
8.
Br J Clin Pharmacol ; 75(2): 476-87, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22759198

ABSTRACT

AIMS: To evaluate apixaban single dose safety, tolerability, pharmacokinetics and pharmacodynamics and assess the effect of food on apixaban pharmacokinetics. METHODS: A double-blind, placebo-controlled, single ascending-dose, first-in-human study assessed apixaban safety, pharmacokinetics and pharmacodynamics in healthy subjects randomized to oral apixaban (n = 43; 0.5-2.5 mg as solution or 5-50 mg as tablets) or placebo (n = 14) under fasted conditions. An open label, randomized, two treatment crossover study investigated apixaban pharmacokinetics/pharmacodynamics in healthy subjects (n = 21) administered apixaban 10 mg in fasted and fed states. Both studies measured apixaban plasma concentration, international normalized ratio (INR), activated partial thromboplastin time (aPTT) and prothrombin time (PT) or a modified PT (mPT). RESULTS: In the single ascending-dose study increases in apixaban exposure appeared dose-proportional. Median t(max) occurred 1.5-3.3 h following oral administration. Mean terminal half-life ranged between 3.6 and 6.8 h following administration of solution doses ≤2.5 mg and between 11.1 and 26.8 h for tablet doses ≥5 mg. Concentration-related changes in pharmacodynamic assessments were observed. After a 50 mg dose, peak aPTT, INR and mPT increased by 1.2-, 1.6- and 2.9-fold, respectively, from baseline. In the food effect study: 90% confidence intervals of geometric mean ratios of apixaban C(max) and AUC in a fed vs. fasted state were within the predefined no effect (80-125%) range. Apixaban half-life was approximately 11.5 h. The effect of apixaban on INR, PT and aPTT was comparable following fed and fasted administration. CONCLUSIONS: Single doses of apixaban were well tolerated with a predictable pharmacokinetic/pharmacodynamic profile and a half-life of approximately 12 h. Apixaban can be administered with or without food.


Subject(s)
Factor Xa Inhibitors , Fibrinolytic Agents/pharmacokinetics , Food-Drug Interactions/physiology , Pyrazoles/pharmacokinetics , Pyridones/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Cross-Over Studies , Double-Blind Method , Fasting , Fibrinolytic Agents/pharmacology , Half-Life , Humans , Male , Pyrazoles/pharmacology , Pyridones/pharmacology , Time Factors , Young Adult
9.
Br J Clin Pharmacol ; 76(3): 432-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23210765

ABSTRACT

AIM(S): This study assessed the effect of differences in renal function on the pharmacokinetics and pharmacodynamics of dapagliflozin, a renal sodium glucose co-transporter-2 (SGLT2) inhibitor for the treatment of type 2 diabetes mellitus (T2DM). METHODS: A single 50 mg dose of dapagliflozin was used to assess pharmacokinetics and pharmacodynamics in five groups: healthy non-diabetic subjects; patients with T2DM and normal kidney function and patients with T2DM and mild, moderate or severe renal impairment based on estimated creatinine clearance. Subsequently, 20 mg once daily multiple doses of dapagliflozin were evaluated in the patients with T2DM. Formation rates of dapagliflozin 3-O-glucuronide (D3OG), an inactive metabolite, were evaluated using human isolated kidney and liver microsomes. RESULTS: Plasma concentrations of dapagliflozin and D3OG were incrementally increased with declining kidney function. Steady-state Cmax for dapagliflozin were 4%, 6% and 9% higher and for D3OG were 20%, 37% and 52% higher in patients with mild, moderate and severe renal impairment, respectively, compared with normal function. AUC(0,τ) was likewise higher. D3OG formation in kidney microsomes was three-fold higher than in liver microsomes and 109-fold higher than in intestine microsomes. Compared with patients with normal renal function, pharmacodynamic effects were attenuated with renal impairment. Steady-state renal glucose clearance was reduced by 42%, 83% and 84% in patients with mild, moderate or severe renal impairment, respectively. CONCLUSIONS: These results indicate that both kidney and liver significantly contribute to dapagliflozin metabolism, resulting in higher systemic exposure with declining kidney function. Dapagliflozin pharmacodynamics in diabetic subjects with moderate to severe renal impairment are consistent with the observation of reduced efficacy in this patient population.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucosides/therapeutic use , Hypoglycemic Agents/therapeutic use , Kidney/physiopathology , Sodium-Glucose Transporter 2 Inhibitors , Adolescent , Adult , Aged , Benzhydryl Compounds , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/urine , Dose-Response Relationship, Drug , Female , Glucosides/adverse effects , Glucosides/pharmacokinetics , Glucosides/pharmacology , Glycosuria/urine , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney Function Tests , Liver/drug effects , Liver/metabolism , Liver/physiopathology , Male , Middle Aged , Sodium-Glucose Transporter 2 , Young Adult
10.
Br J Clin Pharmacol ; 76(5): 776-86, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23451769

ABSTRACT

AIM: Apixaban is an oral factor Xa inhibitor approved for stroke prevention in atrial fibrillation and thromboprophylaxis in patients who have undergone elective hip or knee replacement surgery and under development for treatment of venous thromboembolism. This study examined the safety, pharmacokinetics and pharmacodynamics of multiple dose apixaban. METHOD: This double-blind, randomized, placebo-controlled, parallel group, multiple dose escalation study was conducted in six sequential dose panels - apixaban 2.5, 5, 10 and 25 mg twice daily and 10 and 25 mg once daily- with eight healthy subjects per panel. Within each panel, subjects were randomized (3:1) to oral apixaban or placebo for 7 days. Subjects underwent safety assessments and were monitored for adverse events (AEs). Blood samples were taken to measure apixaban plasma concentration, international normalized ratio (INR), activated partial thromboplastin time (aPTT) and modified prothrombin time (mPT). RESULTS: Forty-eight subjects were randomized and treated (apixaban, n = 36; placebo, n = 12); one subject receiving 2.5 mg twice daily discontinued due to AEs (headache and nausea). No dose limiting AEs were observed. Apixaban maximum plasma concentration was achieved ~3 h post-dose. Exposure increased approximately in proportion to dose. Apixaban steady-state concentrations were reached by day 3, with an accumulation index of 1.3-1.9. Peak : trough ratios were lower for twice daily vs. once daily regimens. Clotting times showed dose-related increases tracking the plasma concentration-time profile. CONCLUSION: Multiple oral doses of apixaban were safe and well tolerated over a 10-fold dose range, with pharmacokinetics with low variability and concentration-related increases in clotting time measures.


Subject(s)
Factor Xa Inhibitors , Fibrinolytic Agents/administration & dosage , Pyrazoles/administration & dosage , Pyridones/administration & dosage , Administration, Oral , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Fibrinolytic Agents/pharmacokinetics , Fibrinolytic Agents/pharmacology , Humans , International Normalized Ratio , Male , Partial Thromboplastin Time , Prothrombin Time , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyridones/pharmacokinetics , Pyridones/pharmacology , Young Adult
11.
Clin Pharmacol Drug Dev ; 12(2): 159-167, 2023 02.
Article in English | MEDLINE | ID: mdl-36369799

ABSTRACT

Pexidartinib is an oral small-molecule tyrosine kinase inhibitor that selectively targets colony-stimulating factor 1 receptor. Two phase 1 single-center trials were conducted in healthy subjects to determine the absorption, distribution, metabolism, and excretion of pexidartinib using radiolabeled drug and to assess the dose proportionality of pexidartinib following single oral doses. In the mass balance study, eight male subjects received a single oral dose of [14 C]-pexidartinib 400 mg with radioactivity assessed in plasma, urine, and feces samples taken at various timepoints postdose. In the dose-proportionality study, 18 subjects received single doses of pexidartinib 200, 400, and 600 mg using randomization sequences. Peak pexidartinib and total radioactivity were observed at 1.75-2.0 hours after the oral dose and then declined in a multiphasic manner. The overall mean recovery of administered radioactivity was 92.2% over 240 hours with 64.8% in the feces and 27.4% in the urine. Major components detected in plasma were pexidartinib and glucuronide (M5, ZAAD-1006a), with M5 and pexidartinib detected in urine and feces, respectively. A glucuronide of dealkylated form (M1) in the urine and multiple oxidized forms (M2, M3, and M4) in feces were detected. The dose-proportionality study found dose-proportional drug exposure between the 200- and 400-mg doses and slightly less than proportional exposure between the 400- and 600-mg doses. These results from these studies provide insight into pexidartinib disposition after oral administration and support the development of dosing guidance in subjects with renal or hepatic impairment or subjects taking cytochrome P450 3A and uridine disphosphate-glucuronosyl transferase inhibitors and inducers.


Subject(s)
Antineoplastic Agents , Glucuronides , Humans , Male , Protein Kinase Inhibitors/pharmacokinetics , Aminopyridines/pharmacokinetics
12.
Clin Pharmacol Drug Dev ; 12(5): 475-483, 2023 05.
Article in English | MEDLINE | ID: mdl-36942508

ABSTRACT

Pexidartinib, an oral small molecule inhibitor of the colony-stimulating factor 1 receptor, is approved for treatment of adults with symptomatic tenosynovial giant cell tumor associated with severe morbidity or functional limitations and not amenable to improvement with surgery. The original dosing regimen is 400 mg of pexidartinib (2 × 200-mg capsules) twice daily, administered on an empty stomach at least 1 hour before or 2 hours after a meal or snack. Because pexidartinib is likely to be taken over an extended period of time, the ability to take pexidartinib with a meal would simplify timing of administration and potentially improve compliance. Since administering 400 mg of pexidartinib with a low-fat meal increases exposure by ≈60% relative to the fasted state, administering 250 mg of pexidartinib with a low-fat meal (low-fat meal dosing regimen) was predicted to achieve an exposure similar to 400 mg administered during a fasted state (original dosing regimen). Based on clinical trial simulations with two one-sided t-tests and bootstrapping (ie, resampling) analyses, a bioequivalence study (n = 24) would have >90% power to conclude that the original dosing regimen (400 mg fasted twice daily) and the low-fat meal dosing regimen (250 mg with a low-fat meal twice daily) are bioequivalent. This report provides the outcome of the implementation of the model-informed drug development strategy to recommend and justify a low-fat meal dosing regimen for pexidartinib that has the potential to improve patient compliance while maintaining drug exposure.


Subject(s)
Aminopyridines , Drug Development , Adult , Humans , Pharmaceutical Preparations , Healthy Volunteers
14.
Clin Pharmacokinet ; 61(11): 1623-1639, 2022 11.
Article in English | MEDLINE | ID: mdl-36264536

ABSTRACT

BACKGROUND AND OBJECTIVE: Pexidartinib is a novel oral small-molecule inhibitor that selectively targets colony-stimulating factor 1 receptor, KIT proto-oncogene receptor tyrosine kinase, and FMS-like tyrosine kinase 3 harboring an internal tandem duplication mutation. It is approved in the United States for the treatment of adult patients with symptomatic tenosynovial giant cell tumor (TGCT) associated with severe morbidity or functional limitations and not amenable to improvement with surgery. Pexidartinib in vitro data indicate the potential for absorption- and metabolism-related drug-drug interactions (DDIs). The objective was to present a comprehensive DDI risk assessment of agents that can impact pexidartinib exposure by altering its absorption and metabolism potentially affecting efficacy and safety of pexidartinib. METHODS: Four open-label crossover studies were performed to assess the effects of a pH modifier (esomeprazole), a strong cytochrome P450 (CYP) 3A4 inhibitor (itraconazole), a strong CYP3A/5'-diphospho-glucuronosyltransferase (UGT) inducer (rifampin), and a UGT inhibitor (probenecid) on the single-dose pharmacokinetics of pexidartinib. In addition, a physiologically based pharmacokinetic model was developed to predict the effect of a moderate CYP3A4 inhibitor (fluconazole) and a moderate CYP3A inducer (efavirenz) on the pharmacokinetics of pexidartinib. RESULTS: Co-administration of pexidartinib with esomeprazole modestly decreased pexidartinib exposure (maximum plasma concentration [Cmax], ng/mL: geometric mean ratio [90% confidence interval (CI)], 45.4% [36.8-55.9]; area under the drug plasma concentration-time curve from time 0 to infinity [AUC∞], ng•h/mL: geometric mean ratio [90% CI], 53.1% [47.4-59.3]), likely related to decreased solubility of pexidartinib at increased pH levels. As expected, the strong CYP3A4 inhibitor itraconazole increased pexidartinib exposure (Cmax, ng/mL: geometric mean ratio [90% CI], 148.3% [127.8-172.0]; AUC∞, ng•h/mL: geometric mean ratio [90% CI], 173.0% [160.7-186.3]) while the strong CYP3A/UGT inducer rifampin decreased exposure (Cmax, ng/mL: geometric mean ratio [90% CI], 67.1% [53.1-84.8]; AUC∞, ng•h/mL: geometric mean ratio [90% CI], 37.0% [30.6-44.8]). In addition, UGT inhibition increased pexidartinib exposure (Cmax, ng/mL: geometric mean ratio [90% CI], 105.8% [92.4-121.0]; AUC∞, ng•h/mL: geometric mean ratio [90% CI], 159.8% [143.4-178.0]), consistent with the fact that pexidartinib is a substrate of the UGT1A4 enzyme, which is responsible for the generation of the major metabolite, ZAAD-1006a. CONCLUSIONS: The physiologically based pharmacokinetic model predicted that a moderate CYP3A4 inhibitor and a moderate CYP3A inducer would produce modest increases and decreases, respectively, in pexidartinib exposure. These results provide a basis for pexidartinib dosing recommendations when administered concomitantly with drugs with drug-drug interaction potential, including dose adjustments when concomitant administration cannot be avoided. CLINICAL TRIAL REGISTRATION: Probenecid: phase I trial, NCT03138759, 3 May, 2017; esomeprazole, itraconazole, rifampin: phase I trials, not registered with ClinicalTrials.gov.


Subject(s)
Cytochrome P-450 CYP3A Inducers , Cytochrome P-450 CYP3A Inhibitors , Adult , Humans , Area Under Curve , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions , Esomeprazole , Healthy Volunteers , Itraconazole , Probenecid , Rifampin
15.
J Clin Pharmacol ; 62(8): 992-1005, 2022 08.
Article in English | MEDLINE | ID: mdl-35247274

ABSTRACT

Pexidartinib is a novel oral small-molecule tyrosine kinase inhibitor targeting the colony-stimulating factor 1 receptor. Pexidartinib undergoes extensive hepatic metabolism via multiple cytochrome P450 and uridine 5'-diphospho-glucuronosyl transferase enzymes, with ZAAD-1006a as the only major metabolite in human plasma. As pexidartinib is extensively metabolized, hepatic impairment (HI) could lead to increased exposure to pexidartinib. The objective of the two phase 1, open-label studies was to determine the pharmacokinetics of pexidartinib after a single 200-mg dose in subjects with mild and moderate HI, based on Child-Pugh classification (PL3397-A-U123: 8 mild HI and 8 moderate HI vs 16 matched healthy controls) and National Cancer Institute Organ Dysfunction Working Group (NCI-ODWG) criteria (PL3397-A-U129: 8 moderate HI versus 8 matched healthy controls [NCT04223635]). Based on Child-Pugh classification, exposure to pexidartinib (maximum observed concentration [Cmax ], area under the plasma concentration-time curve up to the last measurable concentration [AUClast ], and extrapolated to infinity [AUCinf ]) was similar in subjects with mild and moderate HI and in respective matched healthy controls, whereas ZAAD-1006a exposure (AUC) was approximately 27% to 28% and 41% to 48% higher in mild and moderate HI, respectively. According to NCI-ODWG criteria, total pexidartinib exposure was 42% to 46% higher in subjects with moderate HI, compared with healthy controls, and total ZAAD-1006a exposure was 70% to 79% higher for subjects with moderate HI, compared with matched healthy controls with normal hepatic function. These findings were used to develop appropriate dose recommendations in patients with hepatic impairment.


Subject(s)
Liver Diseases , Neoplasms , Aminopyridines , Area Under Curve , Humans , Liver Diseases/metabolism , Multiple Organ Failure , National Cancer Institute (U.S.) , Pyrroles , United States
16.
Chem Res Toxicol ; 24(9): 1575-85, 2011 Sep 19.
Article in English | MEDLINE | ID: mdl-21728364

ABSTRACT

It has recently been proposed that plasma levels of 4ß-hydroxycholesterol (4ßHC) may be indicative of cytochrome P450 3A4 (P450 3A) activity and therefore could be used to probe for P450 3A-mediated drug-drug interactions. With this in mind, we describe a highly sensitive and precise liquid chromatography-electrospray ionization-tandem mass spectrometry method for the measurement of 4ßHC in human plasma with a lower limit of quantification established at 2 ng/mL using 50 µL of plasma. The entire sample preparation scheme including saponification and derivatization of 4ßHC to the corresponding dipicolinyl ester (DPE) was completed in less than 8 h using an automated sample preparation scheme enabling higher-throughput capabilities. Chromatographic resolution of 4ßHC from 4α-hydroxycholesterol and other endogenous isobaric species was achieved in 11-min using an isocratic gradient on a C18 column. Because of endogenous concentrations of 4ßHC in plasma, a stable isotope labeled (SIL) analogue, d7-4ßHC, was used as a surrogate analyte and measured in the standard curve and quality control samples prepared in plasma. A second SIL analogue, d4-4ßHC, was used as the internal standard. The intraday and interday accuracy for the assay was within 6% of nominal concentrations, and the precision for these measurements was less than 5% relative standard deviation. Rigorous stability assessments demonstrated adequate stability of endogenous 4ßHC in plasma and the corresponding DPE derivative for the analysis of clinical study samples. The results from clinical samples following treatment with a potent P450 3A inducer (rifampin) or inhibitor (ketoconazole) are reported and demonstrate the potential future application for this highly precise and robust analytical assay.


Subject(s)
Chromatography, High Pressure Liquid/methods , Hydroxycholesterols/blood , Spectrometry, Mass, Electrospray Ionization/methods , Adult , Chromatography, High Pressure Liquid/economics , Humans , Male , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization/economics , Tandem Mass Spectrometry/economics , Tandem Mass Spectrometry/methods , Time Factors
17.
Br J Clin Pharmacol ; 72(1): 92-102, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21651615

ABSTRACT

AIM: To investigate the effect of co-administration of rifampicin, a potent inducer of cytochrome P450 (CYP) 3A4 enzymes, on the pharmacokinetics (PK) and pharmacodynamics (PD) of saxagliptin and 5-hydroxy saxagliptin in healthy subjects. Saxagliptin is metabolized by CYP3A4/3A5 to 5-hydroxy saxagliptin, its major pharmacologically active metabolite. METHODS: In a non-randomized, open label, single sequence design, 14 healthy subjects received single oral doses of saxagliptin 5 mg with and without steady-state rifampicin (600 mg once daily for 6 days). PK (saxagliptin and 5-hydroxy saxagliptin) and PD (plasma DPP-4 activity) were measured for up to 24 h on days 1 and 7. RESULTS: Concomitant administration with rifampicin resulted in 53% (point estimate 0.47, 90% CI 0.38, 0.57) and 76% (point estimate 0.24, 90% CI 0.21, 0.27) decreases in the geometric mean C(max) and AUC values of saxagliptin, respectively, with a 39% (point estimate 1.39, 90% CI 1.23, 1.56) increase in the geometric mean C(max) and no change (point estimate 1.03, 90% CI 0.97, 1.09) in the AUC of 5-hydroxy saxagliptin. Similar maximum % inhibition and area under the % inhibition-time effect curve over 24 h for DPP-4 activity were observed when saxagliptin was administered alone or with rifampicin. The saxagliptin total active moieties exposure (AUC) decreased by 27% (point estimate 0.73, 90% CI 0.66, 0.81). Saxagliptin with or without rifampicin in this study was generally well tolerated. CONCLUSIONS: Lack of change of PD effect of saxagliptin is consistent with the observed 27% reduction in systemic exposure to the total active moieties, which is not considered clinically meaningful. Based on these findings, it is not necessary to adjust the saxagliptin dose when co-administered with rifampicin.


Subject(s)
Adamantane/analogs & derivatives , Antibiotics, Antitubercular/pharmacology , Dipeptides/pharmacology , Dipeptides/pharmacokinetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Rifampin/pharmacology , Adamantane/pharmacokinetics , Adamantane/pharmacology , Adolescent , Adult , Area Under Curve , Drug Interactions , Female , Humans , Male , Metabolic Clearance Rate/drug effects , Middle Aged , Young Adult
18.
Clin Pharmacol Drug Dev ; 10(9): 974-984, 2021 09.
Article in English | MEDLINE | ID: mdl-34342172

ABSTRACT

This randomized, double-blind, placebo-controlled, ascending single intravenous (IV) bolus-dose study evaluated safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of apixaban, a direct factor Xa (FXa) inhibitor approved for multiple indications. Eight healthy subjects were randomized 3:1 (apixaban:placebo) within each IV dose cohort (0.5, 1.25, 2.5, 3.75, and 5 mg). The 2.5-mg IV panel also received 5 mg of oral apixaban or placebo. Blood samples were collected for PK and PD, including international normalized ratio, modified prothrombin time (mPT), and anti-FXa activity. Apixaban had 66.2% oral bioavailability, dose-proportional exposure, 17 to 26 L steady-state volume of distribution, and 3.2 to 3.5 L/h total plasma clearance. Renal clearance was ≈27%. Anti-FXa activity and mPT changes followed the apixaban plasma concentration-time profile; both were highly correlated with concentration (R2 = 0.99 and R2 = 0.93 for anti-FXa activity and mPT, respectively). International normalized ratio remained within reference range (0.9-1.3). There were no serious or bleeding-related adverse events. Overall, an apixaban single IV bolus was safe and well tolerated over a 10-fold dose range by these subjects. Apixaban had good oral bioavailability, dose-proportional exposure, and constant plasma clearance over a broad dose range, with modest renal clearance. Apixaban PD were consistent with reversible FXa inhibition.


Subject(s)
Factor Xa Inhibitors/administration & dosage , Pyrazoles/administration & dosage , Pyridones/administration & dosage , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Biological Availability , Dose-Response Relationship, Drug , Double-Blind Method , Factor Xa Inhibitors/pharmacokinetics , Factor Xa Inhibitors/pharmacology , Female , Humans , International Normalized Ratio , Male , Prothrombin Time , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyridones/pharmacokinetics , Pyridones/pharmacology , Tissue Distribution , Young Adult
19.
Clin Transl Sci ; 14(6): 2220-2230, 2021 11.
Article in English | MEDLINE | ID: mdl-34080309

ABSTRACT

Milademetan is a small-molecule inhibitor of murine double minute 2 (MDM2) that is in clinical development for advanced solid tumors and hematological cancers, including liposarcoma and acute myeloid leukemia. Milademetan is a CYP3A and P-glycoprotein substrate and moderate CYP3A inhibitor. The current study aims to understand the drug-drug interaction (DDI) risk of milademetan as a CYP3A substrate during its early clinical development. A clinical DDI study of milademetan (NCT03614455) showed that concomitant administration of single-dose milademetan with the strong CYP3A inhibitor itraconazole or posaconazole increased milademetan mean area under the curve from zero to infinity (AUCinf ) by 2.15-fold (90% confidence interval [CI], 1.98-2.34) and 2.49-fold (90% CI, 2.26-2.74), respectively, supporting that the milademetan dose should be reduced by 50% when concomitantly administered with strong CYP3A inhibitors. A physiologically-based pharmacokinetic (PBPK) model of milademetan was subsequently developed to predict the magnitude of CYP3A-mediated DDI potential of milademetan with moderate CYP3A inhibitors. The PBPK model predicted an increase in milademetan exposure of 1.72-fold (90% CI, 1.69-1.76) with fluconazole, 1.91-fold (90% CI, 1.83-1.99) with erythromycin, and 2.02-fold (90% CI, 1.93-2.11) with verapamil. In addition, it estimated that milademetan's original dose (160 mg once daily) could be resumed from its half-reduced dose 3 days after discontinuation of concomitant strong CYP3A inhibitors. The established PBPK model of milademetan was qualified and considered to be robust enough to support continued development of milademetan.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions , Indoles/pharmacokinetics , Pyridines/pharmacokinetics , Pyrrolidines/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Humans , Indoles/administration & dosage , Itraconazole/administration & dosage , Pyridines/administration & dosage , Pyrrolidines/administration & dosage , Risk Assessment , Triazoles/administration & dosage
20.
Clin Pharmacol Ther ; 109(5): 1314-1325, 2021 05.
Article in English | MEDLINE | ID: mdl-33118153

ABSTRACT

Trastuzumab deruxtecan (DS-8201) is a human epidermal growth factor receptor 2 (HER2)-targeting antibody-drug conjugate with a novel enzyme-cleavable linker, a topoisomerase I inhibitor payload, and a drug-to-antibody ratio of ≈ 8. We have characterized the population pharmacokinetics (PK) of trastuzumab deruxtecan and released drug (topoisomerase I inhibitor) in patients with HER2-positive breast cancer or other solid tumor malignancies. This analysis includes pooled data from five clinical studies with 639 patients. Trastuzumab deruxtecan doses ranged from 0.8 to 8.0 mg/kg every 3 weeks. Serum concentrations of trastuzumab deruxtecan and released drug were analyzed using a sequential two-step approach, with the nonlinear mixed-effects modeling methods. Covariate assessment was based upon stepwise forward-addition and backward-elimination process, followed by both univariate and multivariate analysis quantifying their impact on steady-state exposure of trastuzumab deruxtecan and released drug. A two-compartment model with linear elimination best described PK profiles of intact trastuzumab deruxtecan, while a one-compartment model with time-varying release-rate constant and linear elimination described released-drug PK profiles. Statistically significant covariates (country, tumor size, sex, formulation, age, body weight, albumin, total bilirubin, and aspartate aminotransferase) resulted in < 20% change in steady-state area under the concentration-time curve of trastuzumab deruxtecan and released drug, except for increased body weight (95th percentile, 86 kg) and decreased albumin (5th percentile, 31 g/L). Analysis of patients stratified by country, race, renal function, and hepatic function found no clinically meaningful differences in steady-state exposure of intact trastuzumab deruxtecan or released drug. Overall, results suggest that no dose adjustment based on tested covariates or in specific patient populations is warranted.


Subject(s)
Breast Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Immunoconjugates/pharmacokinetics , Trastuzumab/pharmacokinetics , Age Factors , Antineoplastic Agents, Immunological/pharmacokinetics , Body Weight , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Camptothecin/blood , Camptothecin/pharmacokinetics , Drug Liberation , Female , Humans , Immunoconjugates/blood , Male , Models, Biological , Receptor, ErbB-2/metabolism , Trastuzumab/blood
SELECTION OF CITATIONS
SEARCH DETAIL