Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Clin Pharmacol ; 58(6): 717-726, 2018 06.
Article in English | MEDLINE | ID: mdl-29534286

ABSTRACT

Etelcalcetide, a d-amino acid peptide, is an intravenous calcimimetic approved for the treatment of secondary hyperparathyroidism. Etelcalcetide binds the calcium-sensing receptor and increases its sensitivity to extracellular calcium, thereby decreasing secretion of parathyroid hormone (PTH) by chief cells. Etelcalcetide and its low-molecular-weight transformation products are rapidly cleared by renal excretion in healthy subjects, but clearance is substantially reduced and dependent on hemodialysis in end-stage renal disease. The effective half-life is 3-5 days in patients undergoing hemodialysis 3 times a week. A clinical study using a single microtracer intravenous dose of [14 C]etelcalcetide indicated that 60% of the administered dose was eliminated in dialysate. Etelcalcetide undergoes reversible disulfide exchange with serum albumin to form a serum albumin peptide conjugate that is too large (67 kDa) to be dialyzed, until a subsequent exchange forms etelcalcetide or a low-molecular-weight transformation product. This exchange from albumin is apparent after hemodialysis, when it partially restores etelcalcetide concentrations in plasma. Etelcalcetide has no known risks for drug-drug interactions. In phase 3 studies, 74%-75% of hemodialysis patients with secondary hyperparathyroidism who received etelcalcetide achieved a >30% PTH reduction from baseline versus 8%-10% of patients who received placebo. The pharmacokinetics and pharmacodynamics of etelcalcetide in hemodialysis patients supports a 5-mg starting dose administered after hemodialysis and uptitration in 2.5- or 5-mg increments every 4 weeks to a maximum dose of 15 mg 3 times a week.


Subject(s)
Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/metabolism , Peptides/pharmacology , Peptides/pharmacokinetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/therapy , Administration, Intravenous , Calcimimetic Agents/pharmacokinetics , Calcimimetic Agents/pharmacology , Drug Interactions , Humans , Renal Dialysis , Renal Elimination/drug effects , Renal Insufficiency, Chronic/drug therapy
2.
Clin Pharmacokinet ; 57(1): 71-85, 2018 01.
Article in English | MEDLINE | ID: mdl-28508378

ABSTRACT

INTRODUCTION: Etelcalcetide is a novel calcimimetic that binds and activates calcium-sensing receptors (CaSRs) for the treatment of secondary hyperparathyroidism (SHPT). METHODS: To assess titrated dosing regimens, population pharmacokinetic (PK) and PK/pharmacodynamic (PKPD) modeling of etelcalcetide was performed using NONMEM 7.2. In this analysis, plasma etelcalcetide, serum parathyroid hormone (PTH) and calcium (Ca) concentration-time data were collected from five phase I, II, and III clinical trials following single or multiple intravenous doses of etelcalcetide ranging from 2.5 to 60 mg. A semi-mechanistic model was used to describe the relationship between etelcalcetide, PTH, and Ca. This model included the role of PTH in Ca regulation, the feedback of Ca onto PTH production via the CaSR, and the activity of etelcalcetide plasma levels in increasing the sensitivity of the CaSR to Ca via the cooperative binding model. The impact of relevant covariates was evaluated by stepwise forward/backward selection. Model evaluation was based on standard goodness-of-fit plots and prediction-corrected visual predictive checks (pcVPCs). Simulation was conducted to evaluate titrated dosing regimens. RESULTS AND DISCUSSION: The time courses of etelcalcetide, PTH, and Ca were well-described by the model. The clearance and central volume of distribution (Vc) of etelcalcetide were 0.472 L/h and 49.9 L, respectively, while estimates of the turnover half-lives of PTH and Ca were 0.36 and 23 h, respectively. The extent of interindividual variability in model parameters was low to moderate (6-67%), and no covariates were identified as significant predictors of PK and PD variability. pcVPCs confirmed the predictive ability of the model. CONCLUSIONS: The current analysis confirms the putative mechanism of action of etelcalcetide as an allosteric activator of CaSR. Simulations showed that dose titration of etelcalcetide, rather than fixed dose, is needed to effectively decrease the PTH level in patient populations.


Subject(s)
Hyperparathyroidism, Secondary/drug therapy , Models, Biological , Peptides/administration & dosage , Renal Insufficiency, Chronic/therapy , Administration, Intravenous , Adult , Aged , Aged, 80 and over , Calcium/blood , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Male , Middle Aged , Parathyroid Hormone/blood , Peptides/pharmacokinetics , Peptides/pharmacology , Renal Dialysis/methods , Tissue Distribution , Young Adult
3.
J Clin Pharmacol ; 57(10): 1268-1278, 2017 10.
Article in English | MEDLINE | ID: mdl-28513856

ABSTRACT

This study aims at evaluating the utility of the population pharmacokinetics approach in therapeutic protein drug-drug-interaction (DDI) assessment. Simulations were conducted for 2 representative victim drugs, methotrexate and trastuzumab, using a parallel-group design with and without the interaction drug. The effect of a perpetrator on the exposure of the victim drug is described as the ratio of clearance/apparent clearance of the victim drug given with or without the perpetrator. The power of DDI assessment was calculated as the percentage of runs with 90% confidence interval of the estimated DDI effect within 80% to 125% for the scenarios of no DDI, benchmarked with the noncompartmental approach with intensive sampling. The impact of the number of subjects, the number of sampling points per subject, sampling time error, and model misspecification on the power of DDI determination were evaluated. Results showed that with equal numbers of subjects in each arm, the population pharmacokinetics approach with sparse sampling may need about the same or a higher number of subjects compared to a noncompartmental approach in order to achieve similar power. Increasing the number of subjects, even if only in the study drug alone arm, can increase the power. Sampling or dosing time error had notable impacts on the power for methotrexate but not for trastuzumab. Model misspecification had no notable impacts on the power for trastuzumab. Overall, the population pharmacokinetics approach with sparse sampling built in phase 2/3 studies allows appropriate DDI assessment with adequate study design and analysis and can be considered as an alternative to dedicated DDI studies.


Subject(s)
Drug Interactions , Models, Biological , Area Under Curve , Computer Simulation , Humans , Methotrexate/pharmacokinetics , Trastuzumab/pharmacokinetics
4.
Clin Pharmacokinet ; 55(9): 1045-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27056734

ABSTRACT

Romiplostim is a novel thrombopoiesis-stimulating peptibody consisting of a carrier Fc domain and a peptide domain that binds to the thrombopoietin receptor (TPOR) on platelets and platelet precursors. Similar to endogenous thrombopoietin, romiplostim activates the TPOR to stimulate the growth and maturation of megakaryocytes, resulting in increased production of platelets in the circulation. Binding of romiplostim to TPOR on the platelets and megakaryocytes presumably triggers subsequent internalization and degradation. Therefore, increased platelet counts following romiplostim treatment results in increased elimination of the drug. The TPOR target-mediated process is saturable, resulting in nonlinear volume of distribution and clearance of romiplostim. Therefore, target-mediated disposition plays a decreasing role in drug elimination with increasing romiplostim serum concentration. Conversely, nonspecific elimination processes such as renal clearance play an increasing role with increasing romiplostim serum concentration. Limited pharmacokinetics data demonstrated that the exposure to romiplostim was lower after multiple dose administrations than after the first dose, although large inter-subject variability was observed. Large inter- and intra-subject variability in the platelet response was also observed at a given dose. These findings suggest considerable heterogeneity of disease in patients with primary immune thrombocytopenia and support the need for individual dose adjustments based on platelet counts.


Subject(s)
Blood Platelets/drug effects , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Receptors, Fc/therapeutic use , Receptors, Thrombopoietin/drug effects , Recombinant Fusion Proteins/pharmacokinetics , Thrombopoietin/pharmacokinetics , Thrombopoietin/therapeutic use , Animals , Blood Platelets/cytology , Dose-Response Relationship, Drug , Humans , Metabolic Clearance Rate , Mice , Mice, Knockout , Models, Biological , Rats , Receptors, Fc/administration & dosage , Receptors, Fc/blood , Receptors, Fc/drug effects , Receptors, Fc/metabolism , Receptors, Thrombopoietin/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Thrombopoiesis/drug effects , Thrombopoietin/administration & dosage , Thrombopoietin/blood
5.
J Clin Pharmacol ; 55(11): 1236-47, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25951506

ABSTRACT

Data from 3 clinical trials of omecamtiv mecarbil in healthy volunteers and patients with stable heart failure (HF) were analyzed using a nonlinear mixed-effects model to investigate omecamtiv mecarbil's pharmacokinetics and relationship between plasma concentration and systolic ejection time (SET) and Doppler-derived left ventricular outflow tract stroke volume (LVOTSV). Omecamtiv mecarbil pharmacokinetics were described by a linear 2-compartment model with a zero-order input rate for intravenous administration and first-order absorption for oral administration. Oral absorption half-life was 0.62 hours, and absolute bioavailability was estimated as 90%; elimination half-life was approximately 18.5 hours. Variability in pharmacokinetic parameters was not explained by patient baseline characteristics. Omecamtiv mecarbil plasma concentration was directly correlated with increases in SET and LVOTSV between healthy volunteers and patients with HF. The maximum increase from baseline in SET (delta SET) estimated by an Emax model was 137 milliseconds. LVOTSV increased linearly from baseline by 1.6 mL per 100 ng/mL of omecamtiv mecarbil. Model-based simulations for several immediate-release oral dose regimens (37.5, 50, and 62.5 mg dosed every 8, 12, and 24 hours) showed that a pharmacodynamic effect (delta SET ≥20 milliseconds) could be maintained in the absence of excessive omecamtiv mecarbil plasma concentrations.


Subject(s)
Heart Failure/metabolism , Heart Failure/physiopathology , Models, Biological , Urea/analogs & derivatives , Adult , Aged , Cardiac Myosins/metabolism , Cross-Over Studies , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Middle Aged , Stroke Volume/drug effects , Systole/drug effects , Urea/blood , Urea/pharmacokinetics , Urea/pharmacology , Young Adult
6.
J Clin Pharmacol ; 54(5): 593-601, 2014 May.
Article in English | MEDLINE | ID: mdl-24272952

ABSTRACT

Assessment of pharmacokinetic (PK) based drug-drug interactions (DDI) is essential for ensuring patient safety and drug efficacy. With the substantial increase in therapeutic proteins (TP) entering the market and drug development, evaluation of TP-drug interaction (TPDI) has become increasingly important. Unlike for small molecule (e.g., chemical-based) drugs, conducting TPDI studies often presents logistical challenges, while the population PK (PPK) modeling may be a viable approach dealing with the issues. A working group was formed with members from the pharmaceutical industry and the FDA to assess the utility of PPK-based TPDI assessment including study designs, data analysis methods, and implementation strategy. This paper summarizes key issues for consideration as well as a proposed strategy with focuses on (1) PPK approach for exploratory assessment; (2) PPK approach for confirmatory assessment; (3) importance of data quality; (4) implementation strategy; and (5) potential regulatory implications. Advantages and limitations of the approach are also discussed.


Subject(s)
Models, Biological , Proteins/pharmacokinetics , Proteins/therapeutic use , Drug Interactions , Humans
7.
AAPS J ; 15(4): 933-40, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23794076

ABSTRACT

The investigation of therapeutic protein drug-drug interactions has proven to be challenging. In May 2012, a roundtable was held at the American Association of Pharmaceutical Scientists National Biotechnology Conference to discuss the challenges of preclinical assessment and in vitro to in vivo extrapolation of these interactions. Several weeks later, a 2-day workshop co-sponsored by the U.S. Food and Drug Administration and the International Consortium for Innovation and Quality in Pharmaceutical Development was held to facilitate better understanding of the current science, investigative approaches and knowledge gaps in this field. Both meetings focused primarily on drug interactions involving therapeutic proteins that are pro-inflammatory cytokines or cytokine modulators. In this meeting synopsis, we provide highlights from both meetings and summarize observations and recommendations that were developed to reflect the current state of the art thinking, including a four-step risk assessment that could be used to determine the need (or not) for a dedicated clinical pharmacokinetic interaction study.


Subject(s)
Biomedical Research/standards , Biotechnology/standards , Drug Industry/standards , Drug Interactions , United States Food and Drug Administration/standards , Animals , Biomedical Research/trends , Biotechnology/trends , California , Drug Industry/trends , Education/standards , Education/trends , Humans , United States , United States Food and Drug Administration/trends
8.
AAPS J ; 15(1): 172-82, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23139019

ABSTRACT

While therapeutic proteins (TP), particularly recombinant human proteins and fully human monoclonal antibodies, are designed to have a low immunogenic potential in humans, a clinical immune response does sometimes occur and cannot be predicted from preclinical studies. Changes in TP pharmacokinetics may be perceived as an early indication of antibody formation and serve as a surrogate for later changes in efficacy and safety in individual subjects. Given the substantial increase in number of biological products, including biosimilars, there is an urgent need to quantitatively predict and quantify the immune response and any consequential changes in TP pharmacokinetics. The purpose of this communication is to review the utility of population-based modeling and simulation approaches developed to date for investigating the development of an immune response and assessing its impact on TP pharmacokinetics. Two examples of empirical modeling approaches for pharmacokinetic assessment are presented. The first example presents methods to analyze pharmacokinetic data in the presence of anti-drug antibody (ADA) and confirm the effect of immunogenicity on TP pharmacokinetics in early phases of drug development. The second example provides a framework to analyze pharmacokinetic data in the absence or with very low incidence of ADA and confirm with enough power the lack of an immunogenicity effect on TP pharmacokinetics in late phases of drug development. Finally, a theoretical mechanism-based modeling framework is presented to mathematically relate the complex interaction among TP, their targets, and ADA.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Computer Simulation , Humans , Models, Biological
9.
Pharm Res ; 30(3): 655-69, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23250851

ABSTRACT

PURPOSE: Romiplostim is a novel thrombopoiesis-stimulating peptibody that targets the thrombopoietin c-Mpl receptor, resulting in increased platelet production. The pharmacodynamic-mediated disposition (PDMDD) and its stimulatory effect on platelet production in Sprague-Dawley rats, rhesus monkeys, and cynomolgus monkeys following IV bolus and SC administration at various dose levels were determined. METHODS: The pharmacokinetic (PK) profile was described by a PDMDD model that accounts for romiplostim binding to the c-Mpl receptor. The PD model contained a series of aging compartments for precursor cells in bone marrow and platelets. The stimulatory function was described by an on-and-off function operating on the fractional receptor occupancy (RO). The threshold effect, RO(thr), and K(D) parameters were determinants of drug potency, whereas S(max) reflected drug efficacy. RESULTS: The model implicated that receptor-mediated clearance was negligible. RO(thr) estimated occupancies were 0.288, 0.385, 0.771 for rats, rhesus, and cynomolgus monkeys, respectively. The analogous estimated values of K(D) were 4.05, 2320, and 429 ng/mL, implying that romiplostim was much more potent in rats, which was confirmed by a dose-response (ratio of peak platelet count to baseline) relationship. CONCLUSIONS: The model adequately described romiplostim serum concentrations and platelet counts in rats, rhesus monkeys, and cynomolgus monkeys, and quantified linear clearance, PDMDD, and potency of romiplostim.


Subject(s)
Receptors, Thrombopoietin/agonists , Receptors, Thrombopoietin/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/pharmacokinetics , Thrombopoietin/pharmacology , Thrombopoietin/pharmacokinetics , Animals , Blood Platelets/cytology , Blood Platelets/drug effects , Macaca fascicularis , Macaca mulatta , Models, Biological , Platelet Count , Rats , Rats, Sprague-Dawley , Receptors, Fc/blood , Recombinant Fusion Proteins/blood , Thrombopoiesis/drug effects , Thrombopoietin/blood
10.
Clin Pharmacokinet ; 51(4): 247-60, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22420579

ABSTRACT

BACKGROUND AND OBJECTIVE: Denosumab (XGEVA®; AMG 162) is a fully human IgG2 monoclonal antibody, which binds to the receptor activator of nuclear factor κ-B ligand (RANKL) and prevents terminal differentiation, activation and survival of osteoclasts. We aimed to characterize the population pharmacokinetics of denosumab in patients with advanced solid tumours and bone metastases. METHODS: A total of 14 228 free serum concentrations of denosumab from 1076 subjects (495 healthy subjects and 581 advanced cancer patients with solid tumours and bone metastases) included in 14 clinical studies were pooled. Denosumab was administered as either single intravenous (n = 36), single subcutaneous (n = 490) or multiple subcutaneous doses (n = 550) ranging from 30 to 180 mg (or from 0.01 to 3 mg/kg) and was given every 4 or 12 weeks for up to 3 years. An open two-compartment pharmacokinetic model with first-order absorption, linear distribution to a peripheral compartment, linear clearance and quasi-steady-state approximation of the target-mediated drug disposition was used to describe denosumab pharmacokinetics, using NONMEM Version 7.1.0 software. The influence of covariates (body weight, age, race, tumour type) was investigated using the full model approach. Model evaluation was performed through visual predictive checks. Model-based simulations were conducted to explore the role of covariates on denosumab serum concentrations and inferred RANKL occupancy. RESULTS: After subcutaneous administration, the dose-independent bioavailability and mean absorption half-life of denosumab were estimated to be 61% and 2.7 days, respectively. The central volume of distribution and linear clearance were 2.62 L/66 kg and 3.25 mL/h/66 kg, respectively. Clearance and volume parameters were proportional to body weight. Assuming 1 : 1 denosumab-RANKL binding, the baseline RANKL level, quasi-steady-state constant and RANKL degradation rate were inferred to be 4.46 nmol/L, 208 ng/mL and 0.00116 h-1, respectively. Between-subject variability in model parameters was moderate. Following 120 mg dosing every 4 weeks, the inferred RANKL occupancy at steady state exceeded 97% during the entire dosing interval in more than 95% of subjects, regardless of the patient covariates. CONCLUSIONS: The integration of pharmacokinetic data from 14 clinical studies demonstrated denosumab RANKL-mediated pharmacokinetics. Pharmacokinetics-based dosage adjustments on the basis of body weight, age, race and tumour type are not necessary in patients with bone metastases from solid tumours.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Bone Neoplasms/drug therapy , Neoplasms/pathology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Biological Availability , Bone Neoplasms/secondary , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Denosumab , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Male , Models, Biological , RANK Ligand/metabolism , Tissue Distribution
11.
Clin Cancer Res ; 18(9): 2648-57, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22394983

ABSTRACT

PURPOSE: To quantitatively characterize the longitudinal dose exposure-response [urinary N-telopeptide normalized to urinary creatinine (uNTx/Cr) suppression] relationship for denosumab in patients with bone metastases from solid tumors. EXPERIMENTAL DESIGN: Data from 373 patients who received denosumab as single or multiple subcutaneous doses ranging from 30 to 180 mg (or 0.01 to 3 mg/kg) administered every 4 or 12 weeks for up to 3 years were used in this analysis. An inhibitory sigmoid I(Max) model was used to characterize the time course of uNTx/Cr as a function of serum denosumab concentrations and the M3 method was used to analyze the 52% of uNTx/Cr values below the limit of quantification in the context of a mixed-effects model. Age, weight, sex, race, and cancer type were evaluated as potential covariates for model parameters. Model-based simulations were undertaken to explore and predict the role of denosumab dose and dosing intervals on uNTx/Cr suppression. RESULTS: The typical value (between-subject variability; %) for uNTx/Cr at baseline was 49.2 nmol/L/mmol/L (76.8%), denosumab maximal uNTx/Cr suppression (efficacy) was 93.7% (127%), and the denosumab concentration providing half-maximal uNTx/Cr suppression (potency) was 31.8 ng/mL (287%). No effect of covariates on denosumab efficacy and potency was identified. Simulations indicated that a s.c. denosumab dose of 120 mg administered every 4 weeks provides more than 90% suppression of uNTx/Cr in the maximum proportion of patients relative to other every 4- and 12-week doses evaluated. CONCLUSIONS: Over the wide range of dosing regimens examined, a s.c. denosumab dose of 120 mg administered every 4 weeks is the optimal dosing regimen to suppress uNTx/Cr in patients with bone metastases from solid tumors. Clin Cancer Res; 18(9); 2648-57. ©2012 AACR.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Neoplasms/drug therapy , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Bone Neoplasms/secondary , Computer Simulation , Denosumab , Female , Humans , Male , Middle Aged , Models, Statistical , Neoplasms/pathology , RANK Ligand/antagonists & inhibitors , Time Factors
12.
Clin Pharmacokinet ; 50(12): 793-807, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22087866

ABSTRACT

BACKGROUND AND OBJECTIVE: Inhibition of the receptor activator of nuclear factor κ-B ligand (RANKL) is a therapeutic target for treatment of bone disorders associated with increased bone resorption, such as osteoporosis. The objective of this analysis was to characterize the population pharmacokinetics of denosumab (AMG 162; Prolia®), a fully human IgG2 monoclonal antibody that binds to RANKL, in healthy subjects and postmenopausal women with osteopenia or osteoporosis. METHODS: A total of 22944 serum free denosumab concentrations from 495 healthy subjects and 1069 postmenopausal women with osteopenia or osteoporosis were pooled. Denosumab was administered as either a single intravenous dose (n = 36), a single subcutaneous dose (n = 469) or multiple subcutaneous doses (n = 1059), ranging from 0.01 to 3 mg/kg (or 6-210 mg as fixed mass dosages), every 3 or 6 months for up to 48 months. An open, two-compartment pharmacokinetic model with a quasi-steady-state approximation of the target-mediated drug disposition model was used to describe denosumab pharmacokinetics, using NONMEM Version 7.1.0 software. Subcutaneous absorption was characterized by the first-order absorption rate constant (k(a)), with constant absolute bioavailability over the range of doses that were evaluated. Clearance and volume of distribution parameters were scaled by body weight, using a power model. Model evaluation was performed through visual predictive checks. RESULTS: The subcutaneous bioavailability of denosumab was 64%, and the k(a) was 0.00883 h-1. The central volume of distribution and linear clearance were 2.49 L/66 kg and 3.06 mL/h/66 kg, respectively. The baseline RANKL level, quasi-steady-state constant and RANKL degradation rate were 614 ng/mL, 138 ng/mL and 0.00148 h-1, respectively. Between-subject variability in model parameters was moderate. A fixed dose of 60 mg provided RANKL inhibition similar to that achieved by equivalent body weight-based dosing. The effects of age and race on the area under the serum concentration-time curve of denosumab were less than 15% over the range of covariate values that were evaluated. CONCLUSIONS: The non-linearity in denosumab pharmacokinetics is probably due to RANKL binding, and denosumab dose adjustment based on the patient demographics is not warranted.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Bone Density Conservation Agents/pharmacokinetics , Bone Diseases, Metabolic/blood , Models, Biological , Osteoporosis/blood , RANK Ligand/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/blood , Antibodies, Monoclonal, Humanized , Bone Density Conservation Agents/blood , Bone Diseases, Metabolic/drug therapy , Denosumab , Female , Humans , Middle Aged , Osteoporosis/drug therapy , Postmenopause/blood , Young Adult
13.
AAPS J ; 12(4): 729-40, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20963535

ABSTRACT

The objective of this study was to characterize the pharmacokinetics and pharmacodynamics (PK-PD) of romiplostim after single-dose administration in healthy subjects. The mean serum romiplostim concentrations (PK data) and mean platelet counts (PD data) collected from 32 subjects receiving a single intravenous (0.3, 1 and 10 µg/kg) or subcutaneous (0.1, 0.3, 1, and 2 µg/kg) dose were fitted simultaneously to a mechanistic PK-PD model based on pharmacodynamics-mediated drug disposition (PDMDD) and a precursor pool lifespan concept. The two-compartment PK model incorporated receptor-mediated endocytosis and linear mechanisms as parallel elimination pathways. The maximal concentration of receptors (assumed to be proportional to the platelet count), the equilibrium dissociation constant, and the first-order internalization rate constant for endocytosis of the drug-receptor complex were 0.022 fg/platelet, 0.131 ng/mL, and 0.173 h⁻¹, respectively. Romiplostim concentration stimulates the production of platelet precursors via the Hill function, where the SC50 was 0.052 ng/mL and S (max) was 11.2. The estimated precursor cell and platelet lifespans were 5.9 and 10.5 days, respectively. Model-based simulations revealed that the romiplostim exposure and the platelet response are both dependent on the dose administered and the baseline platelet counts. Also, weekly dosing produced a sustained PD response while dosing intervals ≥2 weeks resulted in fluctuating platelet counts. Thus, the mechanistic PK-PD model was suitable for describing the romiplostim PK-PD interplay (PDMDD), the dose-dependent platelet stimulation, and the lifespans of thrombopoietic cell populations.


Subject(s)
Recombinant Fusion Proteins/pharmacology , Thrombopoietin/pharmacology , Adolescent , Adult , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Receptors, Fc , Receptors, Thrombopoietin/agonists , Recombinant Fusion Proteins/pharmacokinetics , Reference Values , Thrombopoietin/pharmacokinetics , Young Adult
14.
J Biopharm Stat ; 20(1): 46-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20077248

ABSTRACT

The European Medicines Agency (EMEA) leads in providing regulatory guidelines to this emerging industry. While biosimilars are not yet available in the United States, as the legal and regulatory pathway is being developed there, biosimilars and other such products are marketed in other regions of the world. We reviewed the European assessment reports of biosimilars to gain insights into the current practices pertaining to the pharmacokinetic (PK) and pharmacodynamic (PD) similarity evaluations because we believe that they might help to shape the future global regulatory landscape. This paper illustrates the molecular complexity of therapeutic proteins, challenges associated with PK and PD evaluations, limitations of similarity assessment using PK and PD endpoints, and design considerations for PK and PD studies. The scope of biotechnology-derived products will be limited to therapeutic protein products, including recombinant human proteins, fusion proteins, and monoclonal antibodies. The challenges to demonstrate similarity are complex and appear to be unique to each therapeutic agent; therefore, the requirements for regulatory approval will most likely continue to be handled at a product-specific level as stipulated in current EMEA guidelines.


Subject(s)
Biological Products/pharmacokinetics , Biomedical Technology/methods , Biomedical Technology/trends , Animals , Biological Products/pharmacology , Biological Products/therapeutic use , Humans , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/trends
16.
J Pharmacokinet Pharmacodyn ; 32(5-6): 767-93, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16328102

ABSTRACT

Pharmacokinetic/pharmacodynamic (PK/PD) models for hematological drug effects exist that assume that cells are produced by a zero- or first-order process, survive for a specific duration (cell lifespan), and then are lost. Due to the fact that delay differential equations (DDE) are needed for cell lifespan models, their software implementation is not straightforward. Our objective is to demonstrate methods to implement three different cell lifespan models for dealing with hematological drug effects and to evaluate the performance of NONMEM to estimate the model parameters. For the basic lifespan indirect response (LIDR) model, cells are produced by a zero-order process and removed due to senescence. The modified LIDR model adds a precursor pool. The LIDR model of cytotoxicity assumes a three-pool indirect model to account for the cell proliferation with capacity-limited cytotoxicity followed by maturation, and removal from the circulation. A numerical method (method of steps) implementing DDE in NONMEM was introduced. Simulation followed by estimation was used to evaluate NONMEM performance and the impact of the minimization algorithm (first-order method vs. first-order conditional estimation method) and the model for residual variability on the estimates of the population parameters. The FOCE method combined with log-transformation of data was found to be superior. This report provides methodology that will assist in application of population methods for assessing hematological responses to various types of drugs.


Subject(s)
Cell Proliferation/drug effects , Models, Biological , Algorithms , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Numerical Analysis, Computer-Assisted , Pharmaceutical Preparations/administration & dosage , Software , Time Factors
17.
J Clin Pharmacol ; 43(4): 406-13, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12723461

ABSTRACT

The objective of this study was to investigate the pharmacokinetics and ex vivo pharmacodynamics of increasing doses of RWJ 67657, along with the effect of food at one dose level in a first-in-human (FIH) study. This was a placebo-controlled, double-blind, randomized trial in healthy male subjects. Subjects received increasing doses of RWJ 67657 or placebo as a single oral dose (0.25-30 mg/kg) under fasting conditions. The effect of food was investigated for the 10-mg/kg dose. Plasma concentrations of RWJ 67657 were measured over a period of 48 hours using a validated LC-MS/MS method. To evaluate the pharmacodynamics of RWJ 67657, inhibition of cytokine production was monitored from exvivo-stimulated polymorphonuclear blood cells (PBMCs). Pharmacokinetic/pharmacodynamic modeling was used to characterize the inhibitory activity of RWJ 67657. RWJ 67657 was rapidly absorbed (mean tmax = 0.6-2.5 h). The pharmacokinetics of RWJ 67657 appear to be nonlinear with respect to single-dose administration of the investigative formulation. Coadministration of food did not have a significant effect on half-life or time to peak concentration (tmax) but decreased the exposure. Mean Cmax values in the presence of food were almost 50% lower than during fasting (542 vs. 1283 ng/mL), and the AUC decreased from 2832 to 1904 ng.h/mL with food. RWJ 67657 inhibited TNF-alpha, IL-8, and IL-6 in a concentration-dependent manner with mean IC50 values of 0.18 microM, 0.04 microM, and 0.43 microM, respectively. At 20 mg/kg, the median inhibition was greater than 85%. There were no significant adverse effects associated with single doses of this drug. This study demonstrates that RWJ 67657 has acceptable safety and pharmacokinetics to warrant further investigation in a repeat-dose setting. In addition, the early determination of effect on biomarkers suggests potential efficacy in diseases mediated by proinflammatory and inflammatory cytokines.


Subject(s)
Imidazoles/pharmacokinetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pyridines/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Biomarkers , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Food-Drug Interactions , Gas Chromatography-Mass Spectrometry , Half-Life , Humans , Hydrogen-Ion Concentration , Imidazoles/administration & dosage , Imidazoles/pharmacology , Male , Pyridines/administration & dosage , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...