Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Pharmacol Exp Ther ; 386(2): 143-155, 2023 08.
Article in English | MEDLINE | ID: mdl-37164370

ABSTRACT

Sutimlimab, a humanized monoclonal antibody targeting the classic complement pathway, is approved in the United States, Japan, and the European Union for the treatment of hemolytic anemia in adults with cold agglutinin disease. The objectives of this study were to support dose selection for phase 3 studies, assess dose recommendations, and establish the relationship between sutimlimab exposure and clinical outcome [hemoglobin (Hb) levels]. Clinically meaningful biomarkers were graphically analyzed and the exposure-response relationship was proposed. The pharmacokinetic (PK) characteristics of sutimlimab were best described by a two-compartment model with parallel linear and nonlinear clearance terms. Body weight was a significant covariate for the volume of distribution in the central compartment (Vc) and total body clearance of sutimlimab. Ethnicity (Japanese, non-Japanese) was a covariate on Vc and maximal nonlinear clearance. There were no PK differences between healthy participants and patients. After graphical exposure-response analysis for biomarkers, a pharmacokinetic-pharmacodynamic model was developed by integrating an indirect response/turnover model for Hb with a maximum effect (Emax) model, relating the Hb-elevating effect of sutimlimab to plasma exposure. Renal function and occurrence of blood transfusion were identified as covariates on Hb change from baseline. Simulations showed that Emax was attained with the approved dosing (6.5 g in patients <75 kg and 7.5 g in patients ≥75 kg), independent of covariate characteristics, and provided adequate sutimlimab exposure to maximize effects on Hb, bilirubin, and total complement component C4 levels. A change in Hb from baseline at steady state of 2.2 g/dl was projected, consistent with phase 3 study observations. SIGNIFICANCE STATEMENT: The final validated population pharmacokinetic (PK) and pharmacokinetic/pharmacodynamic (PK/PD) models confirm that the approved dosing regimen for sutimlimab (6.5 g in patients <75 kg and 7.5 g in patients ≥75 kg) is sufficient, without the need for further dose adjustments in populations of patients with cold agglutinin disease.


Subject(s)
Anemia, Hemolytic, Autoimmune , Adult , Humans , Anemia, Hemolytic, Autoimmune/drug therapy , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers , Body Weight
2.
CPT Pharmacometrics Syst Pharmacol ; 10(11): 1310-1322, 2021 11.
Article in English | MEDLINE | ID: mdl-34478613

ABSTRACT

Writing population analysis reports that fulfilling the specifications of submission readiness is a time-consuming process and prone to human error. Thus, there is a need to streamline the creation of these reports through elements of standardization and automation. LaTeX is considered a suitable program package capable of creating long, modular, structured documents, and, because of its typographic quality, includes formula typesetting. The presented automation scripts together with easily adjustable LaTeX templates are designed to enable the reader to understand and reproduce a typical workflow from analysis to reporting. The focus of this tutorial is to use an example of a population pharmacokinetic analysis to show how to work with the proposed automated structures allowing even a reader new to the concept of LaTeX to automatize the reporting workflow and customize the templates for their specific needs.


Subject(s)
Automation , Humans , Reference Standards , Workflow
4.
J Clin Pharmacol ; 55(7): 815-24, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25683324

ABSTRACT

Cilengitide (EMD 121974, manufactured by Merck KGaA, Darmstadt, Germany) is an αv-integrin receptor antagonist showing high affinity for αvß3 and αvß5.This study determined the mass balance of cilengitide in healthy volunteers receiving a single intravenous infusion of 2.1 MBq (14) C-cilengitide spiked into 250 mL of 2000 mg of cilengitide. Blood, urine, and feces were collected up to day 15 or until excretion of radioactivity was below 1% of the administered dose. Total radioactivity derived from the administration of (14) C-cilengitide and unlabeled cilengitide levels were determined and used for calculation of pharmacokinetic parameters.(14) C-cilengitide-related radioactivity was completely recovered (94.5%; 87.4%-100.6%) and was mainly excreted into urine (mean, 79.0%; range, 70.3%-88.2%) and to a lesser extent into feces (mean, 15.5%; range, 9.3%-20.3%). Of the administered dose, 77.5% was recovered as unchanged cilengitide in urine. The concentration profiles of cilengitide and total radioactivity in plasma were comparable. No circulating metabolites were identified in plasma and urine. Two metabolites,M606-1 and M606-2, were identified in feces considered to be formed by intestinal peptidases or by peptidases from fecal bacteria. In conclusion, the data show that following intravenous administration, (14) C-cilengitide was completely recovered, was excreted mainly via renal elimination, and was not metabolized systemically.


Subject(s)
Integrin alphaVbeta3/antagonists & inhibitors , Receptors, Vitronectin/antagonists & inhibitors , Snake Venoms/pharmacokinetics , Adult , Humans , Infusions, Intravenous , Male , Snake Venoms/administration & dosage , Young Adult
5.
J Clin Pharmacol ; 54(12): 1391-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24911832

ABSTRACT

Cilengitide is an αvß3/αvß5-integrin inhibitor investigated as an anticancer agent. This study aimed to develop a cilengitide population pharmacokinetic model using nonlinear mixed-effects modeling of 136 adult patients with advanced solid tumors and to scale the pharmacokinetic parameters to the pediatric population. A stepwise approach was used, beginning with exploratory analyses checking database/target covariate relationships. A two-compartment structural model was developed to describe cilengitide's concentration-time profile and assess covariates' impact on pharmacokinetic parameters. A bootstrap procedure validated the base/final model stability. A two-compartment model best described concentration-time data. Estimated structural model parameters were: 2.79 L h(-) (1) m(-) (2) central compartment mean systemic clearance, 6.75 L m(-) (2) central compartment volume of distribution, 1.3 L h(-) (1) m(-) (2) intercompartmental clearance, and 3.85 L m(-) (2) peripheral compartment volume of distribution. Mean half-life was 0.9 and 3.8 h (α/ß-phase). Co-medications and study populations had no impact, as the different studies were not significant model covariates. Weight and body surface area correlated with the pharmacokinetic parameters (r = 0.95, P < 0.01). Pharmacokinetic parameters were consistent with individual study-derived parameters; their allometric scaling enabled pediatric pharmacokinetic profile predictions as corroborated by independent data. This model provides the basis for pharmacokinetic profile simulations of different dosages/regimens in different populations.


Subject(s)
Models, Biological , Neoplasms/metabolism , Snake Venoms/pharmacokinetics , Adult , Aged , Aged, 80 and over , Area Under Curve , Body Surface Area , Body Weight , Child , Computer Simulation , Humans , Middle Aged
6.
Invest New Drugs ; 32(2): 347-54, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24242902

ABSTRACT

PURPOSE: We evaluated the safety, tolerability, and pharmacokinetics (PK) of EMD 525797 (DI17E6), a humanized monoclonal antibody targeting αv-integrins, in healthy subjects. METHODS: In this first-in-human, double-blind, placebo-controlled, randomized Phase 1 study, healthy male volunteers were consecutively assigned to 6 ascending single-dose cohorts of 35, 100, 250, 500, 1000, or 1500 mg. Per dose cohort, EMD 525797 or placebo was administered over 1 h as an intravenous 250-mL infusion to 6 and 3 volunteers, respectively. Escalation to the next dose level was based on evaluation of safety, tolerability, and PK data. RESULTS: Fifty-five subjects (aged 18-45 years) were randomized. Twenty-seven of 37 (73 %) subjects receiving EMD 525797 reported a total of 61 adverse events (AEs), including 38 events (in 17 subjects) considered by the investigator to be treatment related. A total of 35 AEs were reported by 14 of 18 (78 %) placebo-treated subjects. The most commonly occurring AEs were gastrointestinal disorders, abnormal laboratory values, and increased or decreased biochemistry and/or hematology values, as well as headaches, which occurred at a slightly higher frequency in the EMD 525797 group compared with placebo. There were no serious AEs or deaths. EMD 525797 PK appeared to be dose dependent, especially at lower doses. CONCLUSION: Ascending single doses of EMD 525797 were shown to be safe and well tolerated. No safety concerns were identified. This study supports the ongoing investigation of EMD 525797.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Integrin alphaV/immunology , Adolescent , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Double-Blind Method , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Middle Aged , Young Adult
7.
AAPS J ; 13(4): 650-61, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22005901

ABSTRACT

Prolonged time delay in response to drug action is a common feature of hematological responses to pharmacotherapy such as erythropoiesis. The objective of this study was to compare the performance of two competing modeling approaches for delayed drug effects, mechanistic cell life span models, and semi-mechanistic cell transit models. The comparison was performed with an experimental dataset from multiple dose administrations of an erythropoietin mimetic to Cynomolgus monkeys. Comparative performance measures include visual predictive checks, goodness-of-fit plots, model estimation time, estimation status, and estimation error. The analysis revealed that both models resulted in a similarly good description of the erythropoietic drug effect, with precision and bias of the model-based predictions of red blood cell counts of less than 11%. The cell transit model needed slightly longer time to converge compared to the cell life span model. The system and drug effect parameters were similar in both models indicating that the models can be interchangeably used to describe the current data. Thus, model selection would be dependent on the purpose of the modeling exercise, the available data, and the time allocated for model development.


Subject(s)
Cellular Senescence , Erythropoiesis/drug effects , Models, Biological , Animals , Hematopoietic Cell Growth Factors/pharmacology , Macaca fascicularis , Pharmacokinetics
8.
BMC Clin Pharmacol ; 10: 14, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-20961422

ABSTRACT

BACKGROUND: Somatropin is recombinant human growth hormone (GH) used for the treatment of growth failure in children and GH deficiency in adults. Two concentrations of a liquid formulation have been developed: 5.83 and 8.0 mg/mL. This trial compared the pharmacokinetics (PK), safety and tolerability of these two liquid concentrations against the freeze-dried (FD) formulation in healthy volunteers. METHODS: In an open-label, single-centre, three-way crossover study, volunteers (aged 18-45 years) were given subcutaneous injections of the reconstituted FD and two liquid formulations in random sequential order, each at 4 mg/dose, with a 1-week wash-out period between doses. To suppress endogenous GH secretion, intravenous somatostatin was infused continuously 1 hour before to 24 hours after each dose, achieving a cumulative dose of 3 mg. Primary PK endpoints were area under the serum concentration-time curve (AUC0-t) and maximum serum concentration (Cmax). For each of the two liquid formulations, bioequivalence with the FD formulation was concluded if the 95% confidence intervals (CIs) for the estimated test/reference ratios of geometric means of AUC0-t and Cmax were within the standard pre-specified acceptance range (0.80-1.25). RESULTS: Fifteen men and 15 women enrolled (safety population, n = 30; PK population, n = 28). Bioequivalence with the FD formulation could be shown for both liquid formulations. The ratios of geometric means (95% CI) were 1.046 (0.980, 1.117) and 0.991 (0.929, 1.058) for AUC0-t and 0.954 (0.875, 1.040) and 0.955 (0.876, 1.041) for Cmax for the 5.83 and 8.0 mg/mL formulations, respectively. No significant differences between the three treatments in half-lives, time to reach Cmax, clearance or volume of distribution were observed. After injection, the most common side-effects were pain or injection-site reactions (all of mild intensity). There were no clinically significant abnormal vital signs, ECG or laboratory findings. There were 56 treatment-related adverse events (AEs): 49 mild, 6 moderate and 1 severe (vomiting). No serious AEs occurred. The pattern of AEs was as expected and all resolved by study end. CONCLUSION: Both concentrations of a new liquid multi-dose formulation are bioequivalent to the FD reference formulation and all are well tolerated. TRIAL REGISTRATION NUMBER: NCT01034735.


Subject(s)
Chemistry, Pharmaceutical/methods , Freeze Drying/methods , Human Growth Hormone/pharmacokinetics , Recombinant Proteins/pharmacokinetics , Therapeutic Equivalency , Administration, Oral , Adolescent , Adult , Biological Availability , Cross-Over Studies , Drug Tolerance , Drug-Related Side Effects and Adverse Reactions , Female , Human Growth Hormone/administration & dosage , Human Growth Hormone/adverse effects , Human Growth Hormone/blood , Humans , Male , Middle Aged , Recombinant Proteins/adverse effects , Young Adult
9.
Clin Pharmacokinet ; 48(7): 477-87, 2009.
Article in English | MEDLINE | ID: mdl-19691369

ABSTRACT

OBJECTIVES: A developed population pharmacokinetic model of the humanized monoclonal antibody (mAb) matuzumab was evaluated by external evaluation. Based on the estimates of the final model, simulations of different dosing regimens and the covariate effect were performed. METHODS: The development dataset included 90 patients, and the evaluation dataset included 81 patients; the two sets of patients were from three different studies. In all studies, the patients had different types of advanced carcinoma - mainly colon, rectal and pancreatic cancer. They received matuzumab as multiple 1-hour intravenous infusions in a wide range of dosing regimens (development dataset: from 400 mg every 3 weeks to 2000 mg in the first week followed by 1600 mg weekly; evaluation dataset: from 100 mg weekly to 800 mg weekly). In addition to 1256 serum mAb concentrations for model development, there were 1124 concentrations available for model evaluation. Serum concentration-time data were simultaneously fitted using NONMEM software. The developed two-compartment model - with the parameters central volume of distribution (V(1)) and peripheral volume of distribution (V(2)), intercompartmental clearance and linear clearance (CLL), an additional nonlinear elimination pathway (Michaelis-Menten constant: the concentration with the half-maximal elimination rate and V(max): the maximum elimination rate) and covariate relations - was evaluated by an external dataset. Different simulation scenarios were performed to demonstrate the impact of the incorporated covariate effect and the influence of different dosing regimens and dosing strategies on the concentration-time profiles. RESULTS: The developed model included the covariate fat-free mass (FFM) on V(1) and on CLL. The evaluation did not support the covariate FFM on V(1) and, after deletion of this covariate, the model parameters of the refined model were estimated. The model showed good precision for all parameters: the relative standard errors (RSEs) were <42% for the development dataset and < or = 51% for the evaluation dataset (excluding the higher RSEs for the correlation between V(2) and V(max) and the interindividual variability on V(2) for the evaluation dataset). The model showed good robustness for the ability to estimate highly precise parameters for the combined dataset of 171 patients (RSE <29%). Simulations revealed that variability in concentration-time profiles for minimum and maximum steady-state concentrations was reduced to a marginal extent by a proposed dose adaptation. CONCLUSION: The population pharmacokinetic model for matuzumab was improved by evaluation with an external dataset. The new model obtained precise parameter estimates and demonstrated robustness. After correlation with efficacy data simulation results in particular could serve as a tool to guide dose selection for this 'targeted' cancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Models, Biological , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Clinical Trials as Topic , Computer Simulation , Data Collection , Drug Administration Schedule , Drug Therapy, Combination , Humans , Infusions, Intravenous , Middle Aged , Neoplasms/drug therapy
10.
J Transl Med ; 7: 68, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19640287

ABSTRACT

BACKGROUND: To explore the biological activity of EMD 273063 (hu14.18-IL2), a humanized anti-GD2 monoclonal antibody fused to interleukin-2 (IL2), in patients with unresectable, stage IV cutaneous melanoma as measured by induction of immune activation at the tumor site and in peripheral blood. METHODS: Nine patients were treated with 4 mg/m2 per day of EMD 273063 given as a 4-h intravenous infusion on days 1, 2, and 3 every four weeks (one cycle). Peripheral blood was analyzed for T cell and natural killer cell phenotype and frequency, as well as levels of soluble IL2 receptor (sIL2R), IL10, IL6, tumor necrosis factor alpha and neopterin. Biopsies of tumor metastasis were performed prior to therapy and at day 10 of the first 2 cycles to study lymphocyte accumulation by immunohistochemistry. RESULTS: Treatment was generally well tolerated and there were no study drug-related grade 4 adverse events. Grade 3 events were mainly those associated with IL2, most commonly rigors (3 patients) and pyrexia (2 patients). Best response on therapy was stable disease in 2 patients. There were no objective tumor regressions by standard response criteria. Systemic immune activation was demonstrated by increases in serum levels of sIL2R, IL10, and neopterin. There was evidence of increased tumor infiltration by T cells, but not NK cells, in most post-dosing biopsies, suggesting recruitment of immune cells to the tumor site. CONCLUSION: EMD 273063 demonstrated biologic activity with increased immune-related cytokines and intratumoral changes in some patients consistent with the suspected mechanism of action of this immunocytokine.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cytokines/therapeutic use , Interleukin-2/therapeutic use , Melanoma/pathology , Melanoma/therapy , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Area Under Curve , Biopsy , Cytokines/administration & dosage , Cytokines/adverse effects , Cytokines/blood , Cytokines/pharmacokinetics , Female , Follow-Up Studies , Half-Life , Humans , Immunohistochemistry , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Interleukin-2/blood , Interleukin-2/pharmacokinetics , Male , Melanoma/surgery , Metabolic Clearance Rate , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Skin Neoplasms/surgery , Time Factors , Treatment Outcome
11.
Clin Toxicol (Phila) ; 46(6): 551-9; discussion 576-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18584369

ABSTRACT

OBJECTIVE: To assess the relationship between blood pressure changes following infusion of antidotal doses of hydroxocobalamin and plasma concentrations of total and free cobalamins-(III). METHODS: Independent groups of healthy volunteers received single intravenous doses of 2.5, 5, 7.5, or 10 g hydroxocobalamin over 7.5 to 30 minutes. RESULTS: In the pharmacokinetic population (n = 41), hydroxocobalamin caused short-lived mean blood pressure increases. Blood pressure increased shortly after initiation of infusion and returned nearly to baseline by 4 hours post-infusion. The time course of blood pressure changes coincided with that of changes in plasma total and free cobalamins-(III). Change in mean arterial pressure (MAP) was strongly correlated with plasma area-under-the-concentration-time curves (AUCs) of total and free cobalamins-(III) during infusion (r > 0.7) but not through 24 hours post-infusion (r < or = 0.36). CONCLUSION: The short-lived increase in mean blood pressure during administration of antidotal doses of hydroxocobalamin is closely linked to initial exposure to total and free cobalamins-(III).


Subject(s)
Antidotes/adverse effects , Blood Pressure/drug effects , Hydroxocobalamin/adverse effects , Vitamin B 12/blood , Adult , Antidotes/administration & dosage , Antidotes/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Female , Humans , Hydroxocobalamin/administration & dosage , Hydroxocobalamin/pharmacokinetics , Infusions, Intravenous , Male , Middle Aged , Poisoning/drug therapy , Time Factors
12.
J Clin Pharmacol ; 48(3): 267-78, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18218786

ABSTRACT

Cetuximab is a monoclonal antibody directed against the epidermal growth factor receptor and is indicated in the treatment of squamous cell carcinoma of the head and neck. The population pharmacokinetics of cetuximab were characterized by nonlinear mixed effects modeling (NONMEM V) using a total of 912 concentrations from 143 patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck enrolled in 2 phase I/II studies. Cetuximab pharmacokinetics were best described by a 2-compartment model with Michaelis-Menten-type saturable elimination. Population estimates (between-subject variability, percent coefficient of variation) of the pharmacokinetic parameters were V(max) 4.38 mg/h (15.4%), K(m) 74 mug/mL, central compartment volume V(1) 2.83 L (18.6%), peripheral compartment volume 2.43 L (56.4%), and intercompartmental clearance 0.103 L/h (97.2%). Ideal body weight and white blood cell count were identified as predictors of V(max) and total body weight as a predictor of V(1). Clinical dose adjustments beyond the approved body surface area-based dosing of cetuximab may be warranted in patients with extreme deviations of their actual body weight from ideal body weight. Agreement between simulated and measured concentrations monitored for up to 43 weeks of therapy indicates that cetuximab pharmacokinetic parameters remained constant during prolonged therapy.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Adult , Aged , Algorithms , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Carcinoma, Squamous Cell/pathology , Cetuximab , Clinical Trials, Phase I as Topic/statistics & numerical data , Clinical Trials, Phase II as Topic/statistics & numerical data , Enzyme-Linked Immunosorbent Assay , Head and Neck Neoplasms/pathology , Humans , Metabolic Clearance Rate , Middle Aged , Models, Biological , Monte Carlo Method , Multicenter Studies as Topic , Neoplasm Metastasis , Randomized Controlled Trials as Topic/statistics & numerical data , Time Factors
13.
Biopharm Drug Dispos ; 28(7): 339-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17654694

ABSTRACT

OBJECTIVE: To investigate the effect of sarizotan on the pharmacokinetics of levodopa in fixed combination with carbidopa or benserazide. METHODS: In this open-label, randomized, crossover study, healthy male subjects (n=16) received levodopa 100 mg t.i.d. over two 5-day periods, alone or in combination with sarizotan 5 mg b.i.d. Levodopa was administered with a dopa-decarboxylase inhibitor (carbidopa 25 mg, n=8 or benserazide 25 mg, n=8). Pharmacokinetic parameters of levodopa were obtained on days 1 and 5. RESULTS: ANOVA showed the C(max) values for levodopa were not significantly different with or without sarizotan after single doses (1001 vs 1082 ng/ml; point estimate [PE] 1.10, 90% confidence intervals [CI] 0.83-1.45) or at steady-state (1549 vs 1663 ng/ml; PE 1.06, 90% CI 0.89-1.27); nor were AUC values for single doses (1661 vs 1665 ng h/ml; PE 1.01, 90% CI 0.91-1.11) or at steady-state (2462 vs 2482 ng h/ml; PE 1.01, 90% CI 0.97-1.05). Seven subjects reported adverse events of mild-to-moderate intensity; the most frequent were headaches and dizziness. CONCLUSION: Coadministration of sarizotan with levodopa, in combination with a dopa-decarboxylase inhibitor had no effect on the pharmacokinetics or adverse event profile of levodopa.


Subject(s)
Levodopa/pharmacokinetics , Adult , Cross-Over Studies , Drug Interactions , Humans , Male , Organic Chemicals/pharmacology
14.
BMC Cancer ; 6: 285, 2006 Dec 11.
Article in English | MEDLINE | ID: mdl-17156477

ABSTRACT

BACKGROUND: Anti-angiogenic treatment is believed to have at least cystostatic effects in highly vascularized tumours like pancreatic cancer. In this study, the treatment effects of the angiogenesis inhibitor Cilengitide and gemcitabine were compared with gemcitabine alone in patients with advanced unresectable pancreatic cancer. METHODS: A multi-national, open-label, controlled, randomized, parallel-group, phase II pilot study was conducted in 20 centers in 7 countries. Cilengitide was administered at 600 mg/m2 twice weekly for 4 weeks per cycle and gemcitabine at 1000 mg/m2 for 3 weeks followed by a week of rest per cycle. The planned treatment period was 6 four-week cycles. The primary endpoint of the study was overall survival and the secondary endpoints were progression-free survival (PFS), response rate, quality of life (QoL), effects on biological markers of disease (CA 19.9) and angiogenesis (vascular endothelial growth factor and basic fibroblast growth factor), and safety. An ancillary study investigated the pharmacokinetics of both drugs in a subset of patients. RESULTS: Eighty-nine patients were randomized. The median overall survival was 6.7 months for Cilengitide and gemcitabine and 7.7 months for gemcitabine alone. The median PFS times were 3.6 months and 3.8 months, respectively. The overall response rates were 17% and 14%, and the tumor growth control rates were 54% and 56%, respectively. Changes in the levels of CA 19.9 went in line with the clinical course of the disease, but no apparent relationships were seen with the biological markers of angiogenesis. QoL and safety evaluations were comparable between treatment groups. Pharmacokinetic studies showed no influence of gemcitabine on the pharmacokinetic parameters of Cilengitide and vice versa. CONCLUSION: There were no clinically important differences observed regarding efficacy, safety and QoL between the groups. The observations lay in the range of other clinical studies in this setting. The combination regimen was well tolerated with no adverse effects on the safety, tolerability and pharmacokinetics of either agent.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Snake Venoms/therapeutic use , Adult , Angiogenesis Inhibitors/toxicity , Antimetabolites, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/toxicity , Cell Division/drug effects , Deoxycytidine/therapeutic use , Deoxycytidine/toxicity , Humans , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Quality of Life , Snake Venoms/toxicity , Surveys and Questionnaires , Survival Rate , Gemcitabine
15.
Clin Toxicol (Phila) ; 44 Suppl 1: 17-28, 2006.
Article in English | MEDLINE | ID: mdl-16990190

ABSTRACT

INTRODUCTION: This randomized, double-blind, placebo-controlled, ascending-dose study was conducted in healthy volunteers to evaluate the safety of the investigational cyanide antidote hydroxocobalamin. METHODS: Four ascending dosing groups received intravenous doses of 2.5, 5, 7.5 or 10 g hydroxocobalamin over 7.5 to 30 minutes at a constant infusion rate. Volunteers (n = 136) randomized 3:1 to receive hydroxocobalamin or placebo underwent a 4-day in-house observation after infusion on Day 1 and follow-up visits on Days 8, 15, and 28. RESULTS: The most common drug-related adverse events were asymptomatic and self-limiting chromaturia and reddening of the skin, which are attributed to the red color of hydroxocobalamin. Other adverse events included pustular/papular rash, headache, erythema at the injection site, decrease in lymphocyte percentage, nausea, pruritus, chest discomfort, and dysphagia. Hydroxocobalamin was associated with an increase in blood pressure in some volunteers. Blood pressure changes peaked toward the end of hydroxocobalamin infusion and typically returned to baseline levels by 4 hours postinfusion. Maximum mean changes from baseline in systolic blood pressure ranged from 22.6 to 27.0 mmHg across hydroxocobalamin doses compared with 0.2 to 6.7 mmHg in the corresponding placebo groups. Maximum mean change from baseline in diastolic blood pressure ranged from 14.3 to 25.4 mmHg across hydroxocobalamin doses compared with -3.0 to 3.8 mmHg in the corresponding placebo groups. Two allergic reactions that occurred within minutes after start of the 5- and 10-g hydroxocobalamin infusions were successfully managed with dexamethasone and/or dimethindene maleate. CONCLUSION: Timely intervention for acute cyanide poisoning could entail administration of an antidote in the prehospital setting based on a presumptive diagnosis. Results of this placebo-controlled study in healthy volunteers corroborate previous studies and French postmarketing experience in cyanide-exposed patients in suggesting that the safety profile of hydroxocobalamin is consistent with prehospital or hospital use.


Subject(s)
Antidotes/adverse effects , Hydroxocobalamin/adverse effects , Adolescent , Adult , Antidotes/administration & dosage , Blood Pressure/drug effects , Cyanides/poisoning , Double-Blind Method , Emergency Medical Services , Female , Heart Rate/drug effects , Humans , Hydroxocobalamin/administration & dosage , Infusions, Intravenous , Male , Middle Aged , Pigments, Biological/urine , Poisoning/drug therapy , Skin/drug effects
16.
Eur J Clin Pharmacol ; 62(4): 277-84, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16525816

ABSTRACT

OBJECTIVE: The 5HT(1A) receptor agonist sarizotan is in clinical development for the treatment of dyskinesia, a potentially disabling complication in Parkinson's disease. We investigated the effect of sarizotan on the clinical pharmacokinetics of probe drugs for cytochrome P450 (CYP) to evaluate the risk of CYP-related drug-drug interactions. METHODS: This was a double-blind, randomised, two-period cross-over interaction study with repeated administration of 5 mg sarizotan HCl or placebo b.i.d. for 8 days in 18 healthy volunteers. On day 4, a single dose of 100 mg metoprolol (CYP2D6 probe) was administered. On day 8, single doses of 100 mg caffeine (CYP1A2 probe), 50 mg diclofenac (CYP2C9 probe), 100 mg mephenytoin (CYP2C19 probe) and 7.5 mg midazolam (CYP3A4 probe) were simultaneously applied. Pharmacokinetic parameters for probe drugs and their metabolites in plasma and urinary recovery were determined. RESULTS: Concentration-time profiles and pharmacokinetic parameters of all probes and their metabolites remained unchanged after co-administration of sarizotan, compared with placebo. Analysis of variance of the area under the plasma concentration-time curve for probe drugs/metabolites, metabolic ratios and urinary excretion resulted in 90% confidence intervals within the acceptance range (0.8-1.25), indicating the absence of drug-drug interactions. CONCLUSIONS: At a dose higher than that intended for clinical use (1 mg b.i.d.), sarizotan had no effect on the metabolism and pharmacokinetics of specific probe drugs for CYP isoenzymes 1A2, 2C19, 2C9, 2D6 and 3A4. Pharmacokinetic interactions with co-administered drugs metabolised by these CYP isoforms are not expected, and dose adjustment of co-administered CYP substrates is not necessary.


Subject(s)
Cytochrome P-450 Enzyme System/analysis , Molecular Probes/pharmacokinetics , Adolescent , Adult , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacology , Aryl Hydrocarbon Hydroxylases/analysis , Cross-Over Studies , Cytochrome P-450 CYP1A2 , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP2D6/analysis , Cytochrome P-450 CYP3A , Double-Blind Method , Drug Interactions , Humans , Isoenzymes/analysis , Male , Mixed Function Oxygenases/analysis , Molecular Probes/administration & dosage , Molecular Probes/metabolism , Organic Chemicals/administration & dosage , Organic Chemicals/pharmacology , Pharmacokinetics
17.
J Clin Pharmacol ; 45(9): 987-1003, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16100293

ABSTRACT

Differences in the clinical pharmacology of the 3 currently available oral phosphodiesterase-5 (PDE5) inhibitors, sildenafil, vardenafil, and tadalafil, are largely determined by their clinical pharmacokinetics as well as their PDE inhibitory activity profile. This review comparatively discusses the major characteristics of the pharmacokinetic profile of all 3 PDE5 inhibitors, including bioavailability and rate of absorption, Biopharmaceutical Classification System categorization, elimination mechanisms, and metabolic profile including active metabolites, as well as the drug-drug interaction potential and modification of pharmacokinetic properties under selected physiologic and pathophysiologic conditions. The review is aimed at providing comparative clinical pharmacology data to allow for scientifically rational, evidence-based prescribing and dosing decisions regarding the clinical use of these medications for the treatment of erectile dysfunction.


Subject(s)
Carbolines/pharmacokinetics , Erectile Dysfunction/drug therapy , Imidazoles/pharmacokinetics , Phosphodiesterase Inhibitors/pharmacokinetics , Piperazines/pharmacokinetics , 3',5'-Cyclic-GMP Phosphodiesterases , Animals , Biological Availability , Cyclic Nucleotide Phosphodiesterases, Type 5 , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Erectile Dysfunction/metabolism , Humans , Male , Metabolic Clearance Rate , Phosphoric Diester Hydrolases/metabolism , Purines , Sildenafil Citrate , Sulfones/pharmacokinetics , Tadalafil , Triazines/pharmacokinetics , Vardenafil Dihydrochloride
18.
Eur J Cancer ; 41(12): 1739-45, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051481

ABSTRACT

This trial assessed pharmacokinetic interactions between cetuximab and irinotecan. Patients were placed in either in group A (irinotecan 350 mg/m2/3 weeks and 400 mg/m2 cetuximab at week 2 then 250 mg/m2/week) or group B (cetuximab weekly starting week 1 then irinotecan starting week 4). Patient plasma or serum samples from each treatment arm were analysed using HPLC and ELISA. Among 14 patients, compartmental model showed no significant differences in mean plasma AUC at week 1 versus week 4 for irinotecan (44,388 versus 39,800 microg/ml/h) and cetuximab (20,441 versus 23,363 microg/ml/h), respectively. Half-lifes (standard deviations) for irinotecan were 16.02 (+/-8.41) h at week 1 and 13.99 (+/-2.14) h at week 4, and for cetuximab 106 (+/-32) at week 3 and 111 (+/-30) h at week 4. Mean concentration-versus-time profiles either alone or in combination were superimposable for cetuximab and irinotecan. From this study, we conclude that there is no evidence of pharmacokinetic interaction between irinotecan and cetuximab.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , ErbB Receptors/metabolism , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cetuximab , Colorectal Neoplasms/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Irinotecan , Male , Middle Aged
19.
J Clin Pharmacol ; 45(6): 631-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15901744

ABSTRACT

Eniporide (EMD 96 875) is a novel and selective inhibitor of the Na+-H+ exchange (NHE-1) inhibitor. The study objectives were to identify a structural model for population pharmacokinetic analysis of eniporide and its metabolite (EMD 112 843) using nonlinear mixed-effects modeling after short-term infusion (dose: 2.5-400 mg) in healthy subjects and patients undergoing myocardial reperfusion therapy. Pooled concentrations of eniporide and its metabolite from healthy subjects (n = 153; 4815 observations) and patients (n = 304; 1465 observations) were included in the pharmacokinetic analysis. Population estimates of clearance and volume of distribution of eniporide were 29.2 L/h (24.1% coefficient of variation [CV], healthy), 20.8 L/h (28.0% CV, patients) and 20.4 L (13.1% CV, healthy), 16.9 L (24.9% CV, patients), respectively. Statistical significance was achieved for the effect of age on clearance and creatinine clearance on volume of distribution of eniporide. The impact of the covariates on eniporide pharmacokinetics is minimal to warrant any dosage adjustments in patient population.


Subject(s)
Guanidines/metabolism , Guanidines/pharmacokinetics , Myocardial Infarction/drug therapy , Sulfones/metabolism , Sulfones/pharmacokinetics , Adult , Analysis of Variance , Female , Guanidines/administration & dosage , Humans , Infusions, Intravenous , Male , Myocardial Reperfusion Injury/drug therapy , Retrospective Studies , Sodium-Hydrogen Exchangers/administration & dosage , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Sulfones/administration & dosage
20.
J Immunother ; 27(3): 232-9, 2004.
Article in English | MEDLINE | ID: mdl-15076141

ABSTRACT

This phase 1 clinical trial was conducted to evaluate the safety and to determine the maximum tolerated dose (MTD) of the immunocytokine EMD 273066 huKS-IL2 and, secondarily, to assess its pharmacokinetics, immunogenic potential, and immunologic activity in patients with androgen-independent prostate cancer (n = 22). EMD 273066 was administered in 3-day cycles (separated by 4 weeks) of once-daily, 4-hour intravenous infusions at a dose determined by an escalation protocol (0.4, 0.7, 1.4, 2.8, 4.3, 6.4, or 8.5 mg/m/d). Approximately 2/3 of patients received a second cycle of treatment. The results show that the MTD of EMD 273066 [ie, one dose level below that producing dose-limiting toxicity (DLT) in at least 33% of patients in a dosing group] was 6.4 mg/m/d. EMD 273066 was generally well tolerated up to a dose of 4.3 mg/m/d. No DLTs, defined as drug-related toxicities >OR= Grade 3 occurring during the first treatment cycle, were observed among patients in the 0.4-, 0.7-, 1.4-, or 4.3-mg/m/d dosing groups. Four patients treated with 2.8, 6.4, or 8.5 mg/m/d EMD 273066 experienced DLTs. Titers of both antiimmunocytokine and anti-FcIL-2 antibody responses were observed after the first dose cycle and either decreased or remained stable during a second course of treatment. No hypersensitivity reactions were observed. EMD 273066 exhibited immunologic activity as demonstrated by increases in lymphocyte counts, natural killer cell number and specific activity, and antibody-dependent cellular cytotoxicity activity. On average, Cmax, which was dose-dependent, was achieved within 1 hour after infusion. Mean t(1/2) which was independent of dose, ranged from 4.0 to 6.7 hours across doses. A zero-compartment body model with one-order kinetics best described the concentration-time profiles. These data demonstrate that the novel immunocytokine EMD 273066 is well tolerated at doses above a level of observed systemic biologic activity in patients with androgen-independent prostate cancer.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Cytokines/therapeutic use , Immunotherapy/methods , Interleukin-2/analogs & derivatives , Interleukin-2/pharmacokinetics , Interleukin-2/therapeutic use , Prostatic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-2/metabolism , Killer Cells, Natural/metabolism , Kinetics , Lymphocytes/drug effects , Male , Maximum Tolerated Dose , Middle Aged , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...