Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Clin Pharmacokinet ; 63(4): 511-527, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38436924

ABSTRACT

BACKGROUND AND OBJECTIVE: The combination of niraparib and abiraterone acetate (AA) plus prednisone is under investigation for the treatment of patients with metastatic castration-resistant prostate cancer (mCRPC) and metastatic castration-sensitive prostate cancer (mCSPC). Regular-strength (RS) and lower-strength (LS) dual-action tablets (DATs), comprising niraparib 100 mg/AA 500 mg and niraparib 50 mg/AA 500 mg, respectively, were developed to reduce pill burden and improve patient experience. A bioequivalence (BE)/bioavailability (BA) study was conducted under modified fasting conditions in patients with mCRPC to support approval of the DATs. METHODS: This open-label randomized BA/BE study (NCT04577833) was conducted at 14 sites in the USA and Europe. The study had a sequential design, including a 21-day screening phase, a pharmacokinetic (PK) assessment phase comprising three periods [namely (1) single-dose with up to 1-week run-in, (2) daily dose on days 1-11, and (3) daily dose on days 12-22], an extension where both niraparib and AA as single-agent combination (SAC; reference) or AA alone was continued from day 23 until discontinuation, and a 30-day follow-up phase. Patients were randomly assigned in a parallel-group design (four-sequence randomization) to receive a single oral dose of niraparib 100 mg/AA 1000 mg as a LS-DAT or SAC in period 1, and patients continued as randomized into a two-way crossover design during periods 2 and 3 where they received niraparib 200 mg/AA 1000 mg once daily as a RS-DAT or SAC. The design was powered on the basis of crossover assessment of RS-DAT versus SAC. During repeated dosing (periods 2 and 3, and extension phase), all patients also received prednisone/prednisolone 5 mg twice daily. Plasma samples were collected for measurement of niraparib and abiraterone plasma concentrations. Statistical assessment of the RS-DAT and LS-DAT versus SAC was performed on log-transformed pharmacokinetic parameters data from periods 2 and 3 (crossover) and from period 1 (parallel), respectively. Additional paired analyses and model-based bioequivalence assessments were conducted to evaluate the similarity between the LS-DAT and SAC. RESULTS: For the RS-DAT versus SAC, the 90% confidence intervals (CI) of geometric mean ratios (GMR) for maximum concentration at a steady state (Cmax,ss) and area under the plasma concentration-time curve from 0-24 h at a steady state (AUC 0-24h,ss) were respectively 99.18-106.12% and 97.91-104.31% for niraparib and 87.59-106.69 and 86.91-100.23% for abiraterone. For the LS-DAT vs SAC, the 90% CI of GMR for AUC0-72h of niraparib was 80.31-101.12% in primary analysis, the 90% CI of GMR for Cmax,ss and AUC 0-24h,ss of abiraterone was 85.41-118.34% and 86.51-121.64% respectively, and 96.4% of simulated LS-DAT versus SAC BE trials met the BE criteria for both niraparib and abiraterone. CONCLUSIONS: The RS-DAT met BE criteria (range 80%-125%) versus SAC based on 90% CI of GMR for Cmax,ss and AUC 0-24h,ss. The LS-DAT was considered BE to SAC on the basis of the niraparib component meeting the BE criteria in the primary analysis for AUC 0-72h; abiraterone meeting the BE criteria in additional paired analyses based on Cmax,ss and AUC 0-24h,ss; and the percentage of simulated LS-DAT versus SAC BE trials meeting the BE criteria for both. GOV IDENTIFIER: NCT04577833.


Subject(s)
Abiraterone Acetate , Indazoles , Piperidines , Prostatic Neoplasms, Castration-Resistant , Tablets , Therapeutic Equivalency , Humans , Indazoles/pharmacokinetics , Indazoles/administration & dosage , Male , Piperidines/pharmacokinetics , Piperidines/administration & dosage , Abiraterone Acetate/pharmacokinetics , Abiraterone Acetate/administration & dosage , Aged , Middle Aged , Prostatic Neoplasms, Castration-Resistant/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Models, Biological , Biological Availability , Cross-Over Studies , Aged, 80 and over , Computer Simulation , Prednisone/pharmacokinetics , Prednisone/administration & dosage
2.
CPT Pharmacometrics Syst Pharmacol ; 13(4): 599-611, 2024 04.
Article in English | MEDLINE | ID: mdl-38298058

ABSTRACT

These analyses characterized tofacitinib pharmacokinetics (PKs) in children and adolescents with juvenile idiopathic arthritis (JIA). Data were pooled from phase I (NCT01513902), phase III (NCT02592434), and open-label, long-term extension (NCT01500551) studies of tofacitinib tablet/solution (weight-based doses administered twice daily [b.i.d.]) in patients with JIA aged 2 to less than 18 years. Population PK modeling used a nonlinear mixed-effects approach, with covariates identified using stepwise forward-inclusion backward-deletion procedures. Simulations were performed to derive dosing recommendations for children and adolescents with JIA. Two hundred forty-six pediatric patients were included in the population PK model. A one-compartment model with first-order elimination and absorption with body weight as a covariate for oral clearance and apparent volume of distribution sufficiently described the data. Oral solution was associated with comparable average concentration (Cavg) and slightly higher (113.9%) maximum concentration (Cmax) versus tablet, which was confirmed by a subsequent randomized, open-label, bioavailability study conducted in healthy adult participants (n = 12) by demonstrating adjusted geometric mean ratios (90% confidence interval) between oral solution and tablet of 1.04 (1.00-1.09) and 1.10 (1.00-1.21) for area under the curve extrapolated to infinity and Cmax, respectively (NCT04111614). A dosing regimen of 3.2 mg b.i.d. solution in patients 10 to less than 20 kg, 4 mg b.i.d. solution in patients 20 to less than 40 kg, and 5 mg b.i.d. tablet/solution in patients greater than or equal to 40 kg, irrespective of age, was proposed to achieve constant Cavg across weight groups. In summary, population PK characterization informed a simplified tofacitinib dosing regimen that has been implemented in pediatric patients with JIA.


Subject(s)
Arthritis, Juvenile , Adult , Humans , Child , Adolescent , Arthritis, Juvenile/drug therapy , Piperidines/pharmacokinetics , Pyrimidines , Tablets
3.
Clin Pharmacol Ther ; 115(3): 422-439, 2024 03.
Article in English | MEDLINE | ID: mdl-38093583

ABSTRACT

Subcutaneous (s.c.) administration of monoclonal antibodies (mAbs) can reduce treatment burden for patients and healthcare systems compared with intravenous (i.v.) infusion through shorter administration times, made possible by convenient, patient-centric devices. A deeper understanding of clinical pharmacology principles related to efficacy and safety of s.c.-administered mAbs over the past decade has streamlined s.c. product development. This review presents learnings from key constituents of the s.c. mAb development pathway, including pharmacology, administration variables, immunogenicity, and delivery devices. Restricted mAb transportation through the hypodermis explains their incomplete absorption at a relatively slow rate (pharmacokinetic (PK)) and may impact mAb-cellular interactions and/or onset and magnitude of physiological responses (pharmacodynamic). Injection volumes, formulation, rate and site of injection, and needle attributes may affect PKs and the occurrence/severity of adverse events like injection-site reactions or pain, with important consequences for treatment adherence. A review of immunogenicity data for numerous compounds reveals that incidence of anti-drug antibodies (ADAs) is generally comparable across i.v. and s.c. routes, and complementary factors including response magnitude (ADA titer), persistence over time, and neutralizing antibody presence are needed to assess clinical impact. Finally, four case studies showcase how s.c. biologics have been clinically developed: (i) by implementation of i.v./s.c. bridging strategies to streamline PD-1/PD-L1 inhibitor development, (ii) through co-development with i.v. presentations for anti-severe acute respiratory syndrome-coronavirus 2 antibodies to support rapid deployment of both formulations, (iii) as the lead route for bispecific T cell engagers (BTCEs) to mitigate BTCE-mediated cytokine release syndrome, and (iv) for pediatric patients in the case of dupilumab.


Subject(s)
Antibodies, Monoclonal , Subcutaneous Tissue , Humans , Child , Antibodies, Monoclonal/adverse effects , Antibodies, Neutralizing , Administration, Intravenous
4.
J Pharmacokinet Pharmacodyn ; 50(6): 475-493, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37925369

ABSTRACT

There are many challenges with rare diseases drug development and rare oncology indications are not different. To understand the regulatory landscape as it relates to application of clinical pharmacology principles in rare oncology product development, we reviewed publicly available information of 39 approvals by US FDA between January 2019 and March 2023. The objective was to understand the expected clinical pharmacology studies and knowledge base in such approvals. Model informed drug development (MIDD) applications were also reviewed, as such approaches are expected to play a critical role in filling clinical pharmacology gaps in rare oncology, where number of clinical trials and size of these trials will perhaps continue to be small. The findings highlighted how clinical pharmacology contributed to the evidence of effectiveness, dose optimization and elucidation of intrinsic and extrinsic factors affecting drug's behavior. Clinical pharmacology studies were often integrated with modeling in many of the NDAs/BLAs. Of the post marketing requirements (PMR) received, 18% were for dose optimization, 49% for DDI, 8% for QTc, 49% for specific population, and 5% for food effect. Two post marketing commitments (PMC) were issued for immunogenicity of the 11 biologics submissions. 15% (6 of 39) of the submissions used maximum tolerated dose (MTD) to advance their molecule into Phase 2 studies. Of them 3 approvals received PMR for dose optimization. 3 + 3 was the most prevalent Phase 1 design with use in 74% of the New Drug Applications (NDA)/Biologic License Applications (BLA) reviewed. Rest used innovative approaches such as BLRM, BOIN or mTPi, with BLRM being the most common. Seamless clinical pharmacology and MIDD approaches are paramount for rare oncology drug development.


Subject(s)
Drug Approval , Pharmacology, Clinical , United States , United States Food and Drug Administration
5.
Cancer Chemother Pharmacol ; 89(4): 499-514, 2022 04.
Article in English | MEDLINE | ID: mdl-35298698

ABSTRACT

PURPOSE: To assess the safety, pharmacokinetics, pharmacodynamics, and preliminary efficacy of cetrelimab (JNJ-63723283), a monoclonal antibody programmed cell death protein-1 (PD-1) inhibitor, in patients with advanced/refractory solid tumors in the phase 1/2 LUC1001 study. METHODS: In phase 1, patients with advanced solid tumors received intravenous cetrelimab 80, 240, 460, or 800 mg every 2 weeks (Q2W) or 480 mg Q4W. In phase 2, patients with melanoma, non-small-cell lung cancer (NSCLC), and microsatellite instability-high (MSI-H)/DNA mismatch repair-deficient colorectal cancer (CRC) received cetrelimab 240 mg Q2W. Response was assessed Q8W until Week 24 and Q12W thereafter. RESULTS: In phase 1, 58 patients received cetrelimab. Two dose-limiting toxicities were reported and two recommended phase 2 doses (RP2D) were defined (240 mg Q2W or 480 mg Q4W). After a first dose, mean maximum serum concentrations (Cmax) ranged from 24.7 to 227.0 µg/mL; median time to Cmax ranged from 2.0 to 3.2 h. Pharmacodynamic effect was maintained throughout the dosing period across doses. In phase 2, 146 patients received cetrelimab 240 mg Q2W. Grade ≥ 3 adverse events (AEs) occurred in 53.9% of patients. Immune-related AEs (any grade) occurred in 35.3% of patients (grade ≥ 3 in 6.9%). Overall response rate was 18.6% across tumor types, 34.3% in NSCLC, 52.6% in programmed death ligand 1-high (≥ 50% by immunohistochemistry) NSCLC, 28.0% in melanoma, and 23.8% in centrally confirmed MSI-H CRC. CONCLUSIONS: The RP2D for cetrelimab was established. Pharmacokinetic/pharmacodynamic characteristics, safety profile, and clinical activity of cetrelimab in immune-sensitive advanced cancers were consistent with known PD-1 inhibitors. TRIAL REGISTRATIONS: NCT02908906 at ClinicalTrials.gov, September 21, 2016; EudraCT 2016-002,017-22 at clinicaltrialsregister.eu, Jan 11, 2017.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Melanoma , Neoplasms , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Apoptosis Regulatory Proteins , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Immune Checkpoint Inhibitors , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Neoplasms/drug therapy , Neoplasms/pathology , Programmed Cell Death 1 Receptor
6.
Clin Pharmacol Drug Dev ; 10(11): 1375-1384, 2021 11.
Article in English | MEDLINE | ID: mdl-34273257

ABSTRACT

Patients may have difficulty swallowing a whole daily dose of 240 mg (4 × 60-mg tablets) of apalutamide. One of the unique properties of apalutamide tablets is easy disintegration and dispersion when mixed into aqueous vehicles, avoiding the need to crush/split the tablets. To evaluate whether this method of apalutamide tablet administration would be conducive in a patient setting, different variations in preparation were evaluated, and one preparation was tested in humans. In vitro compatibility studies evaluated purity, dose, or stability of different variations of apalutamide in applesauce/yogurt/orange juice/green tea. An open-label, randomized, crossover phase 1 study in healthy men determined the bioavailability of an apalutamide-applesauce mixture versus whole tablets based on maximum plasma analyte concentration (Cmax ), area under the plasma analyte concentration-time curve: AUC0-72h and AUC0-168h . Different amounts of applesauce/yogurt/orange juice/green tea as well as durations (up to 6 hours) did not affect the total apalutamide content available. The phase 1 study (n = 12) showed increased total exposure of 5% and peak exposure of 27.6% when comparing the apalutamide-applesauce mixture with whole-tablet administration. Variations in preparation times and total content for applesauce/yogurt/orange juice/green tea did not affect the purity, dose, or stability of apalutamide. An apalutamide-applesauce mixture is a suitable alternative administration method to whole tablets.


Subject(s)
Androgen Antagonists/administration & dosage , Androgen Antagonists/pharmacokinetics , Food , Thiohydantoins/administration & dosage , Thiohydantoins/pharmacokinetics , Administration, Oral , Adult , Biological Availability , Cross-Over Studies , Fruit and Vegetable Juices , Healthy Volunteers , Humans , Male , Malus , Middle Aged , Tablets , Tea , Yogurt
7.
Br J Pharmacol ; 178(19): 3943-3958, 2021 10.
Article in English | MEDLINE | ID: mdl-34008170

ABSTRACT

BACKGROUND AND PURPOSE: Antigen-binding fragment (Fab ) reversal agents were developed to reverse, in bleeding emergency, the long-acting anticoagulant effect of JNJ-64179375 (JNJ-9375), a monoclonal antibody that binds exosite-1 on thrombin. EXPERIMENTAL APPROACH: The pharmacokinetic and pharmacodynamic (PK/PD) activities of three reversal agents of varying in vitro binding affinities to JNJ-9375 were characterised in cynomolgus monkeys. The time course of JNJ-9375 anticoagulant activity and reversal effects of each agent were evaluated. A mechanism-based PK/PD model, which integrated free serum concentrations of reversal agent, total and free serum concentrations of JNJ-9375, and thrombin time, was developed to quantitatively relate JNJ-9375 neutralisation to reversal of induced thrombin time prolongation. Model-based allometric scale-up of the lead reversal agent and the PK/PD relationship of JNJ-9375 in healthy volunteers were utilised to predict clinical dosing regimens. KEY RESULTS: Lowering of free JNJ-9375 by the reversal agents corresponded with reversal of thrombin time prolongation. Total JNJ-9375 displayed typical mAb clearance at 2.75 ml·day-1 ·kg-1 , whereas reversal agents cleared faster between 1400 and 2400 ml·day-1 ·kg-1 . The model-estimated in vivo KD values for JNJ-9375 reversal agents were 9 nM (ICHB-256), 0.4 nM (ICHB-281) and 13.7 pM (ICHB-164), in rank-ordered agreement of their KD values determined in vitro. The three reversal agents exhibited different neutralisation characteristics in vivo, governed primarily by their binding kinetics to JNJ-9375. The model predicted a priori free JNJ-9375 kinetics after dosing ICHB-164 (JNJ-67842125) and JNJ-9375 under a different regimen. CONCLUSION AND IMPLICATIONS: The results enabled selection of JNJ-67842125 as the reversal agent for JNJ-9375.


Subject(s)
Antibodies, Monoclonal , Thrombin , Animals , Antibodies, Monoclonal, Humanized , Blood Coagulation Tests , Macaca fascicularis
8.
Cancer Chemother Pharmacol ; 88(1): 25-37, 2021 07.
Article in English | MEDLINE | ID: mdl-33754187

ABSTRACT

PURPOSE: To assess the safety and pharmacokinetics and determine the recommended phase 2 dose (RP2D) of niraparib with apalutamide or abiraterone acetate plus prednisone (AAP) in patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS: BEDIVERE was a multicenter, open-label, phase 1b study of niraparib 200 or 300 mg/day with apalutamide 240 mg or AAP (abiraterone acetate 1000 mg; prednisone 10 mg). Patients with mCRPC were previously treated with ≥ 2 lines of systemic therapy, including ≥ 1 androgen receptor-axis-targeted therapy for prostate cancer. RESULTS: Thirty-three patients were enrolled (niraparib-apalutamide, 6; niraparib-AAP, 27). No dose-limiting toxicities (DLTs) were reported when combinations included niraparib 200 mg; five patients receiving niraparib 300 mg experienced DLTs [niraparib-apalutamide, 2/3 patients (66.7%); niraparib-AAP, 3/8 patients (37.5%)]. Although data are limited, niraparib exposures were lower when given with apalutamide compared with historical niraparib monotherapy exposures in patients with solid tumors. Because of the higher incidence of DLTs, the niraparib-apalutamide combination and niraparib 300 mg combination with AAP were not further evaluated. Niraparib 200 mg was selected as the RP2D with AAP. Of 19 patients receiving niraparib 200 mg with AAP, 12 (63.2%) had grade 3/4 treatment-emergent adverse events, the most common being thrombocytopenia (26.3%) and hypertension (21.1%). Five patients (26.3%) had adverse events leading to treatment discontinuation. CONCLUSIONS: These results support the choice of niraparib 200 mg as the RP2D with AAP. The niraparib-AAP combination was tolerable in patients with mCRPC, with no new safety signals. An ongoing phase 3 study is further assessing this combination in patients with mCRPC. TRIAL REGISTRATION NO: NCT02924766 (ClinicalTrials.gov).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Indazoles/adverse effects , Indazoles/pharmacokinetics , Piperidines/adverse effects , Piperidines/pharmacokinetics , Poly(ADP-ribose) Polymerase Inhibitors/adverse effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Aged , Aged, 80 and over , Androstenes/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Humans , Indazoles/therapeutic use , Male , Middle Aged , Piperidines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prednisone/therapeutic use , Prostatic Neoplasms, Castration-Resistant/metabolism , Thiohydantoins/therapeutic use
9.
Arthritis Rheumatol ; 71(5): 685-695, 2019 05.
Article in English | MEDLINE | ID: mdl-30427585

ABSTRACT

OBJECTIVE: Tofacitinib is an oral JAK inhibitor for the treatment of rheumatoid arthritis (RA). Altered lymphocyte cell counts and a potential association with increased infection rates have been reported in RA patients treated with JAK inhibitors. This analysis was undertaken to evaluate the short-, mid-, and long-term effects of tofacitinib on lymphocytes and infection rates in patients with RA. METHODS: In this post hoc analysis, absolute lymphocyte counts (ALCs) were obtained from phase III studies (12-24 months; n = 717-958) and phase I/II/III/long-term extension studies of tofacitinib (≤117 months) (All RA population; n = 7,061); lymphocyte subset counts (LSCs) were from phase II studies (1.5-6 months' exposure; n = 236-486), an ORAL Sequel vaccine substudy (~22 months; n = 198), and an ORAL Sequel lymphocyte substudy (~50 months; n = 55-1,035) of tofacitinib. The reversibility of ALC/LSC changes was evaluated. The relationship of ALC and LSC to infections was analyzed in the All RA population. The value of monitoring ALC alone was assessed by examining correlations between ALCs and LSCs. RESULTS: Tofacitinib treatment resulted in an initial increase in ALC versus pretreatment baseline, which gradually declined to steady state by ~48 months. CD4+ and CD8+ T cell counts decreased over long-term treatment, and ALC and LSC changes were reversible upon treatment cessation. Patients with ALCs of <500 cells/mm3 had an increased risk of serious infections. There was no strong association between CD4+ T cell, CD8+ T cell, B cell, or natural killer cell counts and serious infection incidence rates. ALC and CD4+ or CD8+ T cell counts correlated well (R = 0.65-0.86). CONCLUSION: Our findings indicate that monitoring of ALC alone appears to be adequate to assess infection risk in tofacitinib-treated patients with RA.


Subject(s)
Arthritis, Rheumatoid/drug therapy , B-Lymphocytes , Infections/epidemiology , Janus Kinase Inhibitors/therapeutic use , Killer Cells, Natural , Lymphocyte Count , Piperidines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , T-Lymphocytes , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Humans , Incidence , T-Lymphocyte Subsets , Time Factors
10.
Clin Immunol ; 191: 10-20, 2018 06.
Article in English | MEDLINE | ID: mdl-29518577

ABSTRACT

This study evaluated the short-term effects of tofacitinib treatment on peripheral blood leukocyte phenotype and function, and the reversibility of any such effects following treatment withdrawal in healthy volunteers. Cytomegalovirus (CMV)-seropositive subjects received oral tofacitinib 10 mg twice daily for 4 weeks and were followed for 4 weeks after drug withdrawal. There were slight increases in total lymphocyte and total T-cell counts during tofacitinib treatment, and B-cell counts increased by up to 26%. There were no significant changes in granulocyte or monocyte counts, or granulocyte function. Naïve and central memory T-cell counts increased during treatment, while all subsets of activated T cells were decreased by up to 69%. T-cell subsets other than effector memory cluster of differentiation (CD)4+, activated naïve CD4+ and effector CD8+ T-cell counts and B-cell counts, normalized 4 weeks after withdrawal. Following ex vivo activation, measures of CMV-specific T-cell responses, and antigen non-specific T-cell-mediated cytotoxicity and interferon (IFN)-γ production, decreased slightly. These T-cell functional changes were most pronounced at Day 15, partially normalized while still on tofacitinib and returned to baseline after drug withdrawal. Total natural killer (NK)-cell counts decreased by 33%, returning towards baseline after drug withdrawal. NK-cell function decreased during tofacitinib treatment, but without a consistent time course across measured parameters. However, markers of NK-cell-mediated cytotoxicity, antibody-dependent cellular cytotoxicity and IFN-γ production were decreased up to 42% 1 month after drug withdrawal. CMV DNA was not detectable in whole blood, and there were no cases of herpes zoster reactivation. No new safety concerns arose. In conclusion, the effect of short-term tofacitinib treatment on leukocyte composition and function in healthy CMV+ volunteers is modest and largely reversible 4 weeks after withdrawal.


Subject(s)
Janus Kinase Inhibitors/pharmacology , Leukocytes/drug effects , Piperidines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Adult , Aged , Arthritis, Rheumatoid/drug therapy , Female , Healthy Volunteers , Humans , Leukocytes/immunology , Lymphocyte Count , Male , Middle Aged , Phenotype , Piperidines/adverse effects , Pyrimidines/adverse effects , Pyrroles/adverse effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
11.
Br J Clin Pharmacol ; 84(6): 1136-1145, 2018 06.
Article in English | MEDLINE | ID: mdl-29377257

ABSTRACT

AIMS: Tofacitinib is an oral, small molecule JAK inhibitor being investigated for ulcerative colitis (UC). In a phase 2 dose-ranging study, tofacitinib demonstrated efficacy vs. placebo as UC induction therapy. In this posthoc analysis, we aimed to compare tofacitinib dose and plasma concentration as predictors of efficacy and identify covariates that determined efficacy in patients with UC. METHODS: One- and two-compartment pharmacokinetic models, with first-order absorption and elimination, were evaluated to describe plasma tofacitinib concentration-time data at baseline and week 8. Relationships between tofacitinib exposure (dose, average plasma drug concentration during a dosing interval at steady state [Cav,ss ] and trough plasma concentration at steady state [Ctrough,ss ]) and week 8 efficacy endpoints were characterized using logistic regression analysis. Baseline disease, demographics, prior and concurrent UC treatment were evaluated as covariates. RESULTS: Plasma tofacitinib concentrations increased proportionately with dose and estimated oral clearance, and Cav,ss values were not significantly different between baseline and week 8. Dose, Cav,ss and Ctrough,ss performed similarly as predictors of efficacy based on statistical criteria for model fit and comparison of model predictions for each endpoint. Individual Cav,ss values were similar between clinical remitters and nonremitters at predicted efficacious doses (10 and 15 mg twice daily). Baseline Mayo score was a significant determinant of efficacy. Predicted differences from placebo in clinical remission at 10 mg twice daily for patients with baseline Mayo score >8 and ≤8 were 39% (95% CI: 7-70) and 21% (-2-50), respectively. CONCLUSIONS: Exposure-response characterization demonstrated the potential of tofacitinib 10 and 15 mg twice daily as induction therapy for UC without monitoring of plasma drug concentrations for dose optimization.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Colitis, Ulcerative/drug therapy , Gastrointestinal Agents/administration & dosage , Janus Kinase Inhibitors/administration & dosage , Piperidines/administration & dosage , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Adult , Aged , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/pharmacokinetics , Colitis, Ulcerative/blood , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/immunology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Gastrointestinal Agents/adverse effects , Gastrointestinal Agents/blood , Gastrointestinal Agents/pharmacokinetics , Humans , Janus Kinase Inhibitors/adverse effects , Janus Kinase Inhibitors/blood , Janus Kinase Inhibitors/pharmacokinetics , Male , Middle Aged , Models, Biological , Piperidines/adverse effects , Piperidines/blood , Piperidines/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Pyrroles/adverse effects , Pyrroles/blood , Pyrroles/pharmacokinetics , Remission Induction , Severity of Illness Index , Treatment Outcome , Young Adult
12.
Pediatr Rheumatol Online J ; 15(1): 86, 2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29282090

ABSTRACT

BACKGROUND: Juvenile idiopathic arthritis (JIA) is the most common pediatric rheumatic disease and a leading cause of childhood disability. The objective of this study was to characterize the PK, safety, and taste acceptability of tofacitinib in patients with JIA. METHODS: This Phase 1, open-label, multiple-dose (twice daily [BID] for 5 days) study of tofacitinib in patients with active (≥ 5 joints) polyarticular course JIA was conducted from March 2013-December 2015. Patients were allocated to one of three age-based cohorts: Cohort 1, 12 to < 18 years; Cohort 2, 6 to < 12 years; and Cohort 3, 2 to < 6 years. Tofacitinib was administered according to age and body weight as tablets or oral solution (grape flavor). PK were assessed on Day 5; safety was assessed at screening, Day 1, and Day 5. Taste acceptability of the oral solution was evaluated. RESULTS: Twenty-six patients (age range 2-17 years) were enrolled: Cohort 1, N = 8; Cohort 2, N = 9; Cohort 3, N = 9; median tofacitinib doses were 5.0, 2.5, and 3.0 mg BID, respectively. The higher median tofacitinib dose in Cohort 3 versus Cohort 2 reflected implementation of an amended dosing scheme following an interim PK analysis after Cohort 2 recruitment. Geometric mean AUC at steady state (AUCtau) was 156.6 ng•h/mL in Cohort 1, 118.8 ng•h/mL in Cohort 2, and 142.5 ng•h/mL in Cohort 3; Cmax (ng/mL) was 47.0, 41.7, and 66.2, respectively. Ctrough, Cmin, and t1/2 were similar in Cohorts 2 and 3, but higher in Cohort 1. Median time to Cmax (Tmax) was similar between cohorts. Apparent clearance and volume of distribution decreased with decreasing age. Tofacitinib was well tolerated, with no serious adverse events or discontinuations due to adverse events reported. Taste acceptability was confirmed. CONCLUSIONS: PK findings from this study in children with polyarticular course JIA established dosing regimens and acceptable taste for use in subsequent studies within the tofacitinib pediatric development program. TRIAL REGISTRATION: ClinicalTrials.gov: NCT01513902 .


Subject(s)
Arthritis, Juvenile/drug therapy , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics , Administration, Oral , Adolescent , Child , Child, Preschool , Female , Humans , Male , Piperidines/administration & dosage , Piperidines/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrroles/administration & dosage , Pyrroles/adverse effects , Taste , Treatment Outcome
13.
J Clin Pharmacol ; 55(3): 241-50, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25501531

ABSTRACT

Chronic kidney disease (CKD) has been shown to alter the pharmacokinetics of drugs that are eliminated not only via the renal pathway but also by nonrenal clearance and transport. Dosing recommendations in subjects with CKD have historically come from small pharmacokinetic (PK) studies, which have been insulated from the broader clinical development strategy. Opportunities for prospective strategic integration of both preclinical and clinical data on drug clearance mechanisms, model-based approaches, and clinical knowledge of therapeutic index are therefore often missed in designing and analyzing the results of PK studies in subjects with CKD, and eventually providing dosing recommendations. These considerations are valuable in designing informative PK studies in subjects with CKD, as well as for guiding kidney function-related inclusion/exclusion criteria in the broader clinical program and ultimately defining dosing guidelines that optimize benefit-risk balance for these special patient populations based on all available data. This paper offers points to consider for drug developers to increase adoption of a contemporary multidisciplinary approach, which includes key considerations on study design and conduct, methodologies for analysis (population PK and physiologically based PK modeling), and a roadmap to interpret the effect of kidney function on the overall benefit-risk profile of drugs in development.


Subject(s)
Kidney/physiopathology , Pharmaceutical Preparations/administration & dosage , Pharmacokinetics , Renal Insufficiency, Chronic/physiopathology , Animals , Drug Dosage Calculations , Drug Interactions , Humans , Kidney/metabolism , Models, Biological , Renal Elimination , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/metabolism , Risk Factors
14.
Gene Regul Syst Bio ; 2: 1-19, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-19787073

ABSTRACT

Corticosteroids (CS) regulate many enzymes at both mRNA and protein levels. This study used microarrays to broadly assess regulation of various genes related to the greater urea cycle and employs pharmacokinetic/pharmacodynamic (PK/PD) modeling to quantitatively analyze and compare the temporal profiles of these genes during acute and chronic exposure to methylprednisolone (MPL). One group of adrenalectomized male Wistar rats received an intravenous bolus dose (50 mg/kg) of MPL, whereas a second group received MPL by a subcutaneous infusion (Alzet osmotic pumps) at a rate of 0.3 mg/kg/hr for seven days. The rats were sacrificed at various time points over 72 hours (acute) or 168 hours (chronic) and livers were harvested. Total RNA was extracted and Affymetrix gene chips (RG_U34A for acute and RAE 230A for chronic) were used to identify genes regulated by CS. Besides five primary urea cycle enzymes, many other genes related to the urea cycle showed substantial changes in mRNA expression. Some genes that were simply up- or down-regulated after acute MPL showed complex biphasic patterns upon chronic infusion indicating involvement of secondary regulation. For the simplest patterns, indirect response models were used to describe the nuclear steroid-bound receptor mediated increase or decrease in gene transcription (e.g. tyrosine aminotransferase, glucocorticoid receptor). For the biphasic profiles, involvement of a secondary biosignal was assumed (e.g. ornithine decarboxylase, CCAAT/enhancer binding protein) and more complex models were derived. Microarrays were used successfully to explore CS effects on various urea cycle enzyme genes. PD models presented in this report describe testable hypotheses regarding molecular mechanisms and quantitatively characterize the direct or indirect regulation of various genes by CS.

15.
Pharm Res ; 25(4): 769-80, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17674160

ABSTRACT

PURPOSE: This study examines methylprednisolone (MPL) effects on the dynamics of hepatic low-density lipoprotein receptor (LDLR) mRNA and plasma lipids associated with increased risks for atherosclerosis. MATERIALS AND METHODS: Normal male Wistar rats were given 50 mg/kg MPL intramuscularly (IM) and sacrificed at various times. Measurements included plasma MPL and CST, hepatic glucocorticoid receptor (GR) mRNA, cytosolic GR density and hepatic LDLR mRNA, and plasma total cholesterol (TC), low-density lipoprotein cholesterol (LDLC), high density lipoprotein cholesterol (HDLC), and triglycerides (TG). RESULTS: MPL showed bi-exponential disposition with two first-order absorption components. Hepatic GR and LDLR mRNA exhibited circadian patterns which were disrupted by MPL. Down-regulation in GR mRNA (40-50%) was followed by a delayed rebound phase. LDLR mRNA exhibited transient down-regulation (60-70%). Cytosolic GR density was significantly suppressed but returned to baseline by 72 h. Plasma TC and LDLC showed increases (55 and 142%) at 12 h. A mechanistic receptor/gene pharmacokinetic/pharmacodynamic model was developed to describe CS effects on hepatic LDLR mRNA and plasma cholesterols. CONCLUSIONS: Our PK/PD model was able to satisfactorily capture the MPL effects on hepatic LDLR, its relationship to various plasma cholesterols, and builds the foundation to explore this area in the future.


Subject(s)
Glucocorticoids/pharmacology , Lipid Metabolism/drug effects , Lipids/blood , Liver/drug effects , Methylprednisolone Hemisuccinate/pharmacology , Models, Biological , Receptors, LDL/metabolism , Animals , Cholesterol/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Circadian Rhythm , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacokinetics , Injections, Intramuscular , Liver/metabolism , Male , Methylprednisolone Hemisuccinate/administration & dosage , Methylprednisolone Hemisuccinate/pharmacokinetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Receptors, LDL/genetics , Triglycerides/blood
16.
J Pharmacokinet Pharmacodyn ; 34(5): 643-67, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17593325

ABSTRACT

Receptor/gene-mediated effects of corticosteroids on hepatic tyrosine aminotransferase (TAT) were evaluated in normal rats. A group of normal male Wistar rats were injected with 50 mg/kg methylprednisolone (MPL) intramuscularly at the nadir of their plasma corticosterone (CST) rhythm (early light cycle) and sacrificed at various time points up to 96 h post-treatment. Blood and livers were collected to measure plasma MPL, CST, hepatic glucocorticoid receptor (GR) mRNA, cytosolic GR density, TAT mRNA, and TAT activity. The pharmacokinetics of MPL showed bi-exponential disposition with two first-order absorption components from the injection site and bioavailability was 21%. Plasma CST was reduced after MPL dosing, but resumed its daily circadian pattern within 36 h. Cytosolic receptor density was significantly suppressed (90%) and returned to baseline by 72 h resuming its biphasic pattern. Hepatic GR mRNA follows a circadian pattern which was disrupted by MPL and did not return during the study. MPL caused significant down-regulation (50%) in GR mRNA which was followed by a delayed rebound phase (60-70 h). Hepatic TAT mRNA and activity showed up-regulation as a consequence of MPL, and returned to their circadian baseline within 72 and 24 h of treatment. A mechanistic receptor/gene-mediated pharmacokinetic/pharmacodynamic model was able to satisfactorily describe the complex interplay of exogenous and endogenous corticosteroid effects on hepatic GR mRNA, cytosolic free GR, TAT mRNA, and TAT activity in normal rats.


Subject(s)
Corticosterone/physiology , Liver/enzymology , Methylprednisolone/pharmacology , Receptors, Glucocorticoid/physiology , Tyrosine Transaminase/metabolism , Animals , Corticosterone/blood , Male , Methylprednisolone/pharmacokinetics , Models, Biological , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Glucocorticoid/analysis , Receptors, Glucocorticoid/genetics , Tyrosine Transaminase/genetics
17.
Biopharm Drug Dispos ; 28(6): 263-73, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17569107

ABSTRACT

Methylprednisolone (MPL) pharmacokinetics was examined in adrenalectomized (ADX) and normal rats to assess the feasibility of intramuscular (i.m.) dosing for use in pharmacodynamic studies. Several study phases were pursued. Parallel group studies were performed in normal and ADX rats given 50 mg/kg MPL (i.v. or i.m.) and blood samples were collected up to 6 h. Data from studies where normal rats were dosed with 50 mg/kg MPL i.m. and killed over either 6 or 96 h were combined to determine muscle site and plasma MPL concentrations. Lastly, ADX rats were dosed with 50 mg/kg MPL i.m. and killed over 18 h to assess hepatic tyrosine aminotransferase (TAT) dynamics. MPL exhibited bi-exponential kinetics after i.v. dosing with a terminal slope of 2.1 h(-1). The i.m. drug was absorbed slowly with two first-order absorption rate constants, 1.26 and 0.219 h(-1) indicating flip-flop kinetics with overall 50% bioavailability. The kinetics of MPL at the injection site exhibited slow, dual absorption rates. Although i.m. MPL showed lower bioavailability compared with other corticosteroids in rats, TAT dynamics revealed similar i.m. and i.v. response profiles. The more convenient intramuscular dosing can replace the i.v. route without causing marked differences in pharmacodynamics.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Methylprednisolone/pharmacokinetics , Adrenalectomy , Algorithms , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/blood , Area Under Curve , Biological Availability , Half-Life , Injections, Intramuscular , Injections, Intravenous , Liver/metabolism , Male , Metabolic Clearance Rate , Methylprednisolone/administration & dosage , Methylprednisolone/blood , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tyrosine Transaminase/genetics , Tyrosine Transaminase/metabolism
18.
J Pharmacokinet Pharmacodyn ; 34(3): 333-54, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17285360

ABSTRACT

A retrospective analysis was performed to modify our fourth-generation pharmacodynamic model for glucocorticoid receptor (GR) dynamics with incorporation of more physiological features. This modified model was developed by integrating previously reported free cytosolic GR and GR mRNA data following single (10, 50 mg/kg) and dual (50 mg/kg at 0 and 24 hr) intravenous doses of methylprednisolone (MPL) in adrenalectomized (ADX) male Wistar rats with several in vitro studies describing real-time kinetics of the transfer of rat steroid-receptor complex from the cell cytosol to the nucleus. Additionally, free hepatic cytosolic GR and its mRNA data from a chronic infusion dosing study of MPL (0.1 and 0.3 mg/kg/hr) in male ADX Wistar rats were used to verify the predictability of the model. Incorporation of information regarding in vitro receptor kinetics allowed us to describe the receptor-mediated pharmacogenomic effects of MPL for a larger variety of genes in rat liver from microarray studies. These included early responsive gene like CCAAT/enhancer binding protein-beta (CEBP-beta), a transcription factor, as well as the later responsive gene for tyrosine aminotransferase (TAT), a classical biomarker of glucocorticoid (GC) genomic effects. This more mechanistic model of GR dynamics can be applied to characterize profiles for a greater number of genes in liver.


Subject(s)
Cell Nucleus/drug effects , Gene Expression Profiling , Gene Expression/drug effects , Glucocorticoids/pharmacology , Liver/drug effects , Methylprednisolone/pharmacology , Models, Genetic , Receptors, Glucocorticoid/drug effects , Active Transport, Cell Nucleus/drug effects , Adrenalectomy , Animals , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Gene Expression Profiling/methods , Glucocorticoids/administration & dosage , Infusions, Intravenous , Injections, Intravenous , Kinetics , Liver/cytology , Liver/metabolism , Male , Methylprednisolone/administration & dosage , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
19.
J Pharmacokinet Pharmacodyn ; 33(6): 683-717, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17053985

ABSTRACT

Precursor-dependent indirect response (PDIDR) models may describe the tolerance and rebound phenomenon observed for many pharmacodynamic responses where these characteristics are manifested due to the depletion or accumulation of a physiological precursor or cofactor pool responsible for generating drug effects. The purpose of this report is to extend the concepts and applications of these models and to approximate responses and limiting conditions for very large doses of drugs. Asymptotic analysis was performed for qualitative determination of various parameters, such as maximum response (Rmax) and rebound (RBmax), time to maximum response and rebound (TRmax and TRBmax), and area under the effect and rebound curve (ABEC and ABRC) for large doses. Computer simulations were performed to assess the role of dose for both cases where drugs act either by depleting (Model V) or by blocking (Model VI) the endogenous precursor. Simulations showed that Rmax, RBmax, TRBmax, ABEC and ABRC increase with dose, eventually reaching a plateau when Dose/V is very large compared to the efficacy parameters (SC50 or IC50) of the drug. However, TRmax either increased or decreased with dose depending on various system and drug parameters. The limits for these parameters at large doses qualitatively determined by asymptotic analysis closely approximated the plateaus observed from the simulated curves. At large doses, the drug response could be approximated by a Bateman-like function for both Models V and VI. Qualitative analyses along with simulation studies provide a fundamental basis for understanding the temporal aspects of the PDIDR models especially at large doses to describe the tolerance and rebound phenomenon.


Subject(s)
Prodrugs/pharmacology , Area Under Curve , Dose-Response Relationship, Drug , Drug Tolerance , Mathematics , Models, Theoretical , Prodrugs/pharmacokinetics
20.
Eur J Pharm Biopharm ; 54(2): 171-80, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12191689

ABSTRACT

The purpose of this study was to investigate a rapid method for the evaluation of vaporization characteristics for selected benzoic acid derivatives. The compounds studied in this context were the ortho-, meta- and para-derivatives of hydroxy and amino benzoic acids. Calculations for the order of reaction were first carried out for each of the compounds using methyl paraben as the calibration standard. Those compounds undergoing zero order, non-activated evaporation processes, were analyzed by the Antoine and Langmuir equations, conjointly. The coefficient of vaporization was obtained as 1.2 x 10(5)+/-0.8 Pakg (0.5)mol(0.5)s(-1)m(-2)K(-0.5). The vapor pressure values were used to determine the Antoine constants using the SPSS 10.0 software. This study attempts to outline a comprehensive thermogravimetric technique for vapor pressure characterization of single-component systems.


Subject(s)
Benzoates/analysis , Atmospheric Pressure , Chemical Phenomena , Chemistry, Physical , Kinetics , Models, Chemical , Technology, Pharmaceutical/methods , Thermogravimetry/instrumentation , Thermogravimetry/methods , Volatilization
SELECTION OF CITATIONS
SEARCH DETAIL
...