Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Eur J Drug Metab Pharmacokinet ; 49(1): 71-85, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38044419

ABSTRACT

BACKGROUND: Overactive adenosine triphosphate signaling via P2X3 homotrimeric receptors is implicated in multiple conditions. To fully understand the metabolism and elimination pathways of eliapixant, a study was conducted to assess the pharmacokinetics, mass balance, and routes of excretion of a single oral dose of the selective P2X3 receptor antagonist eliapixant, in addition to an in vitro characterization. METHODS: In this single-center open-label non-randomized non-placebo-controlled phase I study, healthy male subjects (n = 6) received a single dose of 50 mg eliapixant blended with 3.7 MBq [14C]eliapixant as a PEG 400-based oral solution. Total radioactivity and metabolites excreted in urine and feces, and pharmacokinetics of total radioactivity, eliapixant, and metabolites in plasma were assessed via liquid scintillation counting and high-performance liquid chromatography-based methods coupled to radiometric and mass spectrometric detection. Metabolite profiles of eliapixant in human in vitro systems and metabolizing enzymes were also investigated. RESULTS: After administration as an oral solution, eliapixant was rapidly absorbed, reaching maximum plasma concentrations within 2 h. Eliapixant was eliminated from plasma with a mean terminal half-life of 48.3 h. Unchanged eliapixant was the predominant component in plasma (72.6% of total radioactivity area under the curve). The remaining percentage of drug-related components in plasma probably represented the sum of many metabolites, detected in trace amounts. Mean recovery of total radioactivity was 97.9% of the administered dose (94.3-99.4%) within 14 days, with 86.3% (84.8-88.1%) excreted via feces and 11.6% (9.5-13.1%) via urine. Excretion of parent drug was minimal in feces (0.7% of dose) and urine (≈ 0.5%). In feces, metabolites formed by oxidation represented > 90% of excreted total radioactivity. The metabolites detected in the in vitro experiments were similar to those identified in vivo. CONCLUSION: Complete recovery of administered eliapixant-related radioactivity was observed in healthy male subjects with predominant excretion via feces. Eliapixant was almost exclusively cleared by oxidative biotransformation (> 90% of dose), with major involvement of cytochrome P450 3A4. Excretion of parent drug was of minor importance (~ 1% of dose). CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov: NCT04487431 (registered 27 July 2020)/EudraCT number: 2020-000519-54 (registered 3 February 2020), NCT02817100 (registered 26 June 2016), NCT03310645 (registered 16 October 2017).


Eliapixant is a drug that acts on structures in the body called P2X3 receptors that are involved in several conditions, including chronic cough, overactive bladder, and endometriosis-related pain. When evaluating a new drug, it is important to know how it is being removed from the body by natural mechanisms. We performed a study in which six healthy male volunteers took a single dose of eliapixant, and we investigated what happened to the drug after it was taken. We measured the amount of eliapixant in the volunteers' blood, urine, and feces, and also measured the compounds formed when eliapixant was broken down naturally by the body ("metabolites"). We also used human cells in the laboratory to investigate how the different metabolites of eliapixant are formed. Almost three-quarters of eliapixant in the blood had not been broken down at all, while the remaining one-quarter had been converted into many different metabolites. A total of 2 weeks after taking eliapixant, almost all of it had been converted to metabolites and eliminated from the body (mostly in feces, but also a small amount in urine). The most important organ for breaking down eliapixant is the liver. The information from this study will help doctors determine whether eliapixant is likely to interfere with other drugs taken simultaneously, and whether patients with liver or kidney problems might take longer than healthy people to remove it from their bodies.


Subject(s)
Metabolic Networks and Pathways , Purinergic P2X Receptor Antagonists , Humans , Male , Chromatography, High Pressure Liquid/methods , Mass Spectrometry , Feces/chemistry , Administration, Oral , Volunteers , Healthy Volunteers
2.
Clin Pharmacol Ther ; 105(4): 1031-1039, 2019 04.
Article in English | MEDLINE | ID: mdl-30402880

ABSTRACT

M281 is a fully human, anti-neonatal Fc receptor (FcRn) antibody that inhibits FcRn-mediated immunoglobulin G (IgG) recycling to decrease pathogenic IgG while preserving IgG production. A randomized, double-blind, placebo-controlled, first-in-human study with 50 normal healthy volunteers was designed to probe safety and the physiological maximum for reduction of IgG. Intravenous infusion of single ascending doses up to 60 mg/kg induced dose-dependent serum IgG reductions, which were similar across all IgG subclasses. Multiple weekly doses of 15 or 30 mg/kg achieved mean IgG reductions of ≈85% from baseline and maintained IgG reductions ≥75% from baseline for up to 24 days. M281 was well tolerated, with no serious or severe adverse events (AEs), few moderate AEs, and a low incidence of infection-related AEs similar to placebo treatment. The tolerability and consistency of M281 pharmacokinetics and pharmacodynamics support further evaluation of M281 in diseases mediated by pathogenic IgG.


Subject(s)
Antibodies/metabolism , Antibodies/therapeutic use , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Receptors, Fc/metabolism , Adult , Antibodies/adverse effects , Double-Blind Method , Female , Healthy Volunteers , Humans , Infusions, Intravenous/methods , Male , Young Adult
3.
Invest New Drugs ; 37(4): 711-721, 2019 08.
Article in English | MEDLINE | ID: mdl-30569245

ABSTRACT

Adenosine suppresses antitumor immune responses via A2a and A2b receptors expressed on intratumoral immune cells. This effect is mediated by increased cyclic adenosine 5'-monophosphate (AMP) levels and phosphorylation of cyclic AMP response element binding protein (CREB). We conducted a phase 1, placebo-controlled, single-ascending-dose (SAD) and multiple-ascending-dose (MAD) study to assess the safety, tolerability, pharmacokinetics (PK), including food effect (FE), and pharmacodynamics (PD) of oral AB928, a novel dual A2aR/A2bR antagonist, in healthy volunteers. AB928 doses between 10 and 200 mg once daily and 100 mg twice daily were evaluated. The study enrolled 85 subjects (randomized 3:1, AB928:placebo), 40 each in the SAD and MAD cohorts, and 5 in the FE cohort. AB928 was well tolerated up to the highest dose tested and did not affect any physiologic parameters potentially sensitive to adenosine inhibition. No safety concern was identified. The PK profile of AB928 was linear and dose-proportional, and a clear PK/PD correlation was demonstrated. Significant inhibition of adenosine receptor-mediated phosphorylated CREB was observed at peak plasma concentrations in all dose cohorts and at trough plasma concentrations in the higher-dose cohorts. AB928 plasma levels ≥1 µM were associated with ≥90% adenosine receptor inhibition. In the postprandial state, the rate of AB928 absorption decreased but the extent of absorption was unchanged. Together, these data support further clinical development of oral AB928 in cancer patients.


Subject(s)
Purinergic P1 Receptor Antagonists/administration & dosage , Administration, Oral , Adolescent , Adult , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Double-Blind Method , Female , Food-Drug Interactions , Healthy Volunteers , Humans , Male , Middle Aged , Purinergic P1 Receptor Antagonists/blood , Purinergic P1 Receptor Antagonists/pharmacokinetics , Young Adult
4.
Rheumatol Ther ; 5(2): 403-421, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29959660

ABSTRACT

INTRODUCTION: BI 695501 has shown similar efficacy, safety, and immunogenicity to the adalimumab reference product, Humira®. We present two phase 1 studies comparing the pharmacokinetics, safety, and immunogenicity of BI 695501 delivered via autoinjector (AI) vs. prefilled syringe (PFS). METHODS: Both trials were randomized, open-label, parallel-group studies undertaken in subjects aged ≥ 18-65 years. VOLTAIRE®-AI (NCT02606903) recruited healthy, Caucasian, male, non-athletic volunteers with BMI ≥ 18 to ≤ 30 kg/m2. VOLTAIRE®-TAI (NCT02899338) recruited healthy men and women with BMI > 17.5 to < 35 kg/m2. In both studies, a single dose of BI 695501 40 mg was administered via AI or PFS to the abdomen (VOLTAIRE®-AI) or thigh (VOLTAIRE®-TAI). The observation period was 43/57 days and the safety follow-up was 70 days. Co-primary endpoints were AUC0-1032 or AUC0-1368, Cmax, and AUC0-∞. Safety and immunogenicity were assessed. RESULTS: Subjects (VOLTAIRE®-AI: N = 71; VOLTAIRE®-TAI: N = 162) were randomized to AI (n = 35; n = 81) or PFS (n = 36; n = 81). Baseline characteristics were balanced between treatment groups in each study. Total exposure of BI 695501 was similar for both groups; adjusted geometric mean ratios for AUC0-∞, AUC0-1032, and Cmax were 106.17, 104.09, and 114.83%, respectively, for VOLTAIRE®-AI; 103.19, 101.71 (AUC0-1368), and 100.11% for VOLTAIRE®-TAI. In both studies, similar immunogenicity was observed between groups in terms of frequency of binding and neutralizing anti-drug antibody-positive subjects. Incidence of adverse events was similar for both groups. CONCLUSIONS: Pharmacokinetics and immunogenicity of BI 695501 delivered via AI were similar to administration using a PFS, independent of injection site. No differences are expected between AI and PFS use in clinical practice. FUNDING: Boehringer Ingelheim.

5.
J Cancer Res Clin Oncol ; 144(6): 1087-1095, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29671069

ABSTRACT

PURPOSE: Pegfilgrastim is a long-acting granulocyte colony-stimulating factor indicated for prevention of febrile neutropenia in patients receiving myelosuppressive chemotherapy by promoting neutrophil recovery. METHODS: This phase 1, randomized, double-blind, three-way crossover trial in healthy volunteers evaluated the pharmacokinetics (PK), pharmacodynamics (PD), safety, and tolerability of the proposed biosimilar, comparing MYL-1401H, reference pegfilgrastim (Neulasta®, Amgen Inc, Thousand Oaks, CA, USA) sourced from the European Union, and reference pegfilgrastim sourced from the USA. Primary PK end points were peak plasma concentration of pegfilgrastim (Cmax) and area under the plasma concentration-time curve from the time of dosing to infinity (AUC0-inf). Primary PD end points were area under the curve above baseline for absolute neutrophil counts (ANC AUC0-t) and maximum change from baseline for ANC (ANC Cmax). Adverse events were also recorded. RESULTS: The primary PK and PD end points were similar across all groups. For the PK parameters, the 90% confidence intervals (CIs) of the ratios of geometric means ranged between 0.91 and 1.18, which were within the predefined bioequivalence interval of 0.8000 to 1.2500 for all comparisons. For the PD parameters, the 95% CIs of the ratios of geometric means ranged between 0.94 and 1.06 for all comparisons, which were within the predefined PD equivalence interval of 0.8500 to 1.1765. The safety profiles were similar, with the most common adverse events being back pain and headache. CONCLUSIONS: MYL-1401H demonstrated similar PK, PD, and safety to reference pegfilgrastim in healthy volunteers and may be an equivalent option for the prevention of febrile neutropenia.


Subject(s)
Biosimilar Pharmaceuticals/pharmacology , Biosimilar Pharmaceuticals/pharmacokinetics , Filgrastim/pharmacology , Filgrastim/pharmacokinetics , Polyethylene Glycols/pharmacology , Polyethylene Glycols/pharmacokinetics , Adult , Biosimilar Pharmaceuticals/adverse effects , Chemotherapy-Induced Febrile Neutropenia/blood , Chemotherapy-Induced Febrile Neutropenia/etiology , Cross-Over Studies , Double-Blind Method , Female , Filgrastim/adverse effects , Humans , Male , Polyethylene Glycols/adverse effects , Therapeutic Equivalency
6.
BMC Gastroenterol ; 18(1): 3, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29304731

ABSTRACT

BACKGROUND: Pathogenesis in non-alcoholic steatohepatitis (NASH) involves abnormal cholesterol metabolism and hepatic accumulation of toxic free cholesterol. Apical sodium-dependent bile acid transporter (ASBT) inhibition in the terminal ileum may facilitate removal of free cholesterol from the liver by reducing recirculation of bile acids (BAs) to the liver, thereby stimulating new BA synthesis from cholesterol. The aim of this phase 1 study in adult healthy volunteers (HVs) and patients with type 2 diabetes mellitus (T2DM) was to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of ASBT inhibition with volixibat (SHP626; formerly LUM002). METHODS: Participants were randomised 3:1 to receive once-daily oral volixibat (0.5 mg, 1 mg, 5 mg or 10 mg) or placebo for 28 days in two cohorts (HV and T2DM). Assessments included safety, faecal BA and serum 7α-hydroxy-4-cholesten-3-one (C4; BA synthesis biomarker). RESULTS: Sixty-one individuals were randomised (HVs: placebo, n = 12; volixibat, n = 38; T2DM: placebo, n = 3; volixibat, n = 8). No deaths or treatment-related serious adverse events were reported. Mild or moderate gastrointestinal adverse events were those most frequently reported with volixibat. With volixibat, mean total faecal BA excretion on day 28 was ~1.6-3.2 times higher in HVs (643.73-1239.3 µmol/24 h) and ~8 times higher in T2DM (1786.0 µmol/24 h) than with placebo (HVs: 386.93 µmol/24 h; T2DM: 220.00 µmol/24 h). With volixibat, mean C4 concentrations increased by ~1.3-5.3-fold from baseline to day 28 in HVs and by twofold in T2DM. CONCLUSIONS: Volixibat was generally well tolerated. Increased faecal BA excretion and serum C4 levels support the mechanistic rationale for exploring ASBT inhibition in NASH. The study was registered with the Dutch clinical trial authority (Centrale Commissie Mensgebonden Onderzoek; trial registration number NL44732.056.13; registered 24 May 2013).


Subject(s)
Benzothiepins/administration & dosage , Benzothiepins/adverse effects , Diabetes Mellitus, Type 2/drug therapy , Glycosides/administration & dosage , Glycosides/adverse effects , Membrane Glycoproteins/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/drug therapy , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors , Adolescent , Adult , Aged , Benzothiepins/pharmacokinetics , Bile Acids and Salts/analysis , Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Cholestenones/blood , Diabetes Mellitus, Type 2/metabolism , Double-Blind Method , Feces/chemistry , Female , Glycosides/pharmacokinetics , Homeostasis , Humans , Lipid Metabolism , Male , Middle Aged , Young Adult
7.
Sci Transl Med ; 9(387)2017 04 26.
Article in English | MEDLINE | ID: mdl-28446688

ABSTRACT

Conventional cytotoxic chemotherapy is highly effective in certain cancers but causes dose-limiting damage to normal proliferating cells, especially hematopoietic stem and progenitor cells (HSPCs). Serial exposure to cytotoxics causes a long-term hematopoietic compromise ("exhaustion"), which limits the use of chemotherapy and success of cancer therapy. We show that the coadministration of G1T28 (trilaciclib), which is a small-molecule inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6), contemporaneously with cytotoxic chemotherapy protects murine hematopoietic stem cells (HSCs) from chemotherapy-induced exhaustion in a serial 5-fluorouracil treatment model. Consistent with a cell-intrinsic effect, we show directly preserved HSC function resulting in a more rapid recovery of peripheral blood counts, enhanced serial transplantation capacity, and reduced myeloid skewing. When administered to healthy human volunteers, G1T28 demonstrated excellent in vivo pharmacology and transiently inhibited bone marrow (BM) HSPC proliferation. These findings suggest that the combination of CDK4/6 inhibitors with cytotoxic chemotherapy should provide a means to attenuate therapy-induced BM exhaustion in patients with cancer.


Subject(s)
Hematopoietic Stem Cells/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Female , Fluorouracil/pharmacology , Healthy Volunteers , Hematopoietic Stem Cells/cytology , Humans , Male , Mice , Mice, Inbred C57BL
8.
Clin Pharmacokinet ; 55(10): 1227-1237, 2016 10.
Article in English | MEDLINE | ID: mdl-27147514

ABSTRACT

BACKGROUND AND OBJECTIVE: Previous clinical trials have suggested that bovine intestinal alkaline phosphatase has renal protective effects in patients with sepsis-associated acute kidney injury. We conducted a first-in-human study to investigate the pharmacokinetics, safety and tolerability of a novel human recombinant alkaline phosphatase (recAP), and we developed a population pharmacokinetic model to support dose selection for future patient studies. METHODS: In a randomized, double-blind, placebo-controlled, phase I trial, healthy volunteers received a single dose of recAP (200, 500, 1000 or 2000 U/kg; n = 33; 3:1 ratio) or multiple doses of recAP (500 or 1000 U/kg; n = 18; 2:1 ratio) via a 1-h intravenous infusion on three consecutive days. Serum recAP concentrations, alkaline phosphatase (AP) activity levels and anti-drug antibodies were measured, and safety parameters were monitored. A population pharmacokinetic model was developed, and simulations were performed to guide dose selection for a phase IIa/b trial. RESULTS: Peak concentrations of recAP and peak AP activity were reached at the end of the 1-h infusion and showed a rapid decline, with about 10 % of the maximum concentration remaining at 4 h and less than 5 % remaining 24 h post-start. RecAP treatment was generally well tolerated, and anti-drug antibodies could not be detected in the serum up to 2 weeks post-injection after a single dose, or up to 3 weeks post-injection after multiple doses. A four-compartment model best described the pharmacokinetics of recAP administration, with moderate inter-individual variability on the central volume of distribution and elimination rate constant. Simulations showed that 1-h intravenous infusions of 250, 500 and 1000 U/kg recAP once every 24 h for three consecutive days constituted the dosing regimen that best met the criteria for dose selection in patient studies. CONCLUSION: RecAP did not raise any safety concerns when administered to healthy volunteers. A population pharmacokinetic model was developed to support dose selection for patient studies. TRIAL REGISTRATION: 2013-002694-21 (EudraCT).


Subject(s)
Alkaline Phosphatase/pharmacokinetics , Adolescent , Adult , Body Mass Index , Computer Simulation , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Middle Aged , Models, Biological , Racial Groups , Recombinant Proteins , Young Adult
9.
Clin Ther ; 37(2): 311-24, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25438724

ABSTRACT

PURPOSE: Encenicline (EVP-6124) is a selective α7 nicotinic acetylcholine receptor partial agonist being developed for cognitive impairment in Alzheimer's disease and schizophrenia. We report on 2 single-dose studies to assess the relative bioavailability, pharmacokinetic profile, tolerability, and cognitive effects of encenicline in healthy volunteers. METHODS: A single ascending-dose study assessed the safety, tolerability, pharmacokinetic, and pharmacodynamic profiles of encenicline in healthy male volunteers. Subjects received a single 1-, 3.5-, 7-, 20-, 60-, or 180-mg oral solution dose of encenicline or placebo. A second single-dose, randomized, open-label, 3-period, crossover study in healthy male and female subjects compared the relative bioavailability of a 1-mg oral capsule versus a 1-mg oral solution dose of encenicline and evaluated the effects of food and sex on encenicline pharmacokinetic profile. FINDINGS: In the first study, encenicline was well tolerated and dose-proportional increases in C(max) (mean range 0.59-100 ng/mL) and AUC0-∞ (mean range 45.6-8890 ng·h/mL) were observed over a 1- to 180-mg dose range. Procognitive effects on the Digit Symbol Substitution Test were maximal at the 20-mg dose. In the second study, encenicline 1-mg oral capsules and oral solution were bioequivalent and there was no observed food effect on encenicline pharmacokinetic profile with the 90% confidence intervals of the treatment ratios for both comparisons (ie, capsule to solution and fed to fasted) for Cmax and AUC being within 80% to 125%. A 30% to 40% higher encenicline exposure in female subjects than respective values in male subjects was consistent with a 33% higher weight of the male subjects. No clinically relevant safety profile or tolerability effects of encenicline were observed. IMPLICATIONS: Encenicline was well tolerated at single doses up to 180 mg, and doses as low as 1 mg had dose- and time-dependent pharmacodynamic effects on the central nervous system. Oral capsule and solution were bioequivalent and were not affected by food. Although a sex effect on pharmacokinetic profile was observed, it was attributable to weight differences. Clinical Trial Registration at EudraCT: 2006-005623-42 and EudracT: 2008-000029-20.


Subject(s)
Nicotinic Agonists/pharmacokinetics , Quinuclidines/pharmacokinetics , Thiophenes/pharmacokinetics , Administration, Oral , Adult , Biological Availability , Capsules , Cognition/drug effects , Cross-Over Studies , Double-Blind Method , Fasting , Female , Food-Drug Interactions , Healthy Volunteers , Humans , Male , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/adverse effects , Quinuclidines/administration & dosage , Quinuclidines/adverse effects , Receptors, Nicotinic , Sex Factors , Therapeutic Equivalency , Thiophenes/administration & dosage , Thiophenes/adverse effects
10.
Antivir Ther ; 18(7): 931-40, 2013.
Article in English | MEDLINE | ID: mdl-23963204

ABSTRACT

BACKGROUND: Approximately one-third of all HIV-infected individuals are coinfected with HCV, many of whom will receive concomitant treatment for both infections. With the advent of direct-acting antivirals (DAAs) for HCV, potential drug interactions between antiretrovirals and DAAs require evaluation prior to co-therapy. METHODS: Three open-label studies were conducted in healthy subjects to assess potential interactions between the investigational first-in-class HCV NS5A replication complex inhibitor daclatasvir and representative antiretrovirals atazanavir/ritonavir, efavirenz and tenofovir disoproxil fumarate. RESULTS: Target exposure was that of 60 mg daclatasvir alone. Dose-normalized (60 mg) geometric mean ratios of daclatasvir AUCτ for 20 mg ± atazanavir/ritonavir (2.10 [90% CI 1.95, 2.26]) and 120 mg ± efavirenz (0.68 [0.60, 0.78]) showed less than the three-fold elevation and two-fold reduction, respectively, in systemic exposure predicted by prior interaction studies with potent inhibitors/inducers of CYP3A4. Daclatasvir dose adjustment to 30 mg once daily with atazanavir/ritonavir and 90 mg once daily with efavirenz is predicted to normalize AUCτ relative to the target exposure (geometric mean ratios 1.05 [0.98, 1.13] and 1.03 [0.90, 1.16], respectively). Atazanavir exposure (Cmax, AUCτ and C24 trough) and efavirenz Ctrough under coadministration were similar to historical data without daclatasvir. No clinically relevant interactions between daclatasvir and tenofovir disoproxil fumarate were observed for either drug, and no dosing adjustments were indicated. Daclatasvir was well tolerated in all three studies. CONCLUSIONS: The pharmacokinetic data support coadministration of daclatasvir with atazanavir/ritonavir, efavirenz and/or tenofovir. A Phase III study in HIV-HCV coinfection has commenced using the described dose modifications.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Viral Nonstructural Proteins/antagonists & inhibitors , Adenine/administration & dosage , Adenine/analogs & derivatives , Adenine/pharmacokinetics , Adolescent , Adult , Alkynes , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Atazanavir Sulfate , Benzoxazines/administration & dosage , Benzoxazines/pharmacokinetics , Carbamates , Coinfection , Cyclopropanes , Drug Interactions , Female , HIV Infections/drug therapy , Hepatitis C/drug therapy , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Male , Middle Aged , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Organophosphonates/administration & dosage , Organophosphonates/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Pyrrolidines , Ritonavir/administration & dosage , Ritonavir/pharmacokinetics , Tenofovir , Valine/analogs & derivatives , Young Adult
11.
Br J Clin Pharmacol ; 75(5): 1221-30, 2013 May.
Article in English | MEDLINE | ID: mdl-22978318

ABSTRACT

AIM: Conjugation to antithrombin III ATIII-binding pentasaccharides has been proposed as a novel method to extend the half-life of therapeutic proteins. We aim to validate this technological concept in man by performing a first-in-human study using CarboCarrier® insulin (SCH 900948) as an example. A rising single dose phase 1 study was performed assessing safety, tolerability, pharmacokinetics and relative bioactivity of CarboCarrier® insulin. Safety, tolerability and pharmacokinetics (PK) of single doses of CarboCarrier® insulin in healthy volunteers were explored, and the dose-response relationship and relative bioactivity of CarboCarrier® insulin in subjects with type 2 diabetes were investigated. METHODS: After an overnight fast, subjects were randomized to a treatment sequence. PK and pharmacodynamic (glucose, insulin and C-peptide) samples were obtained for up to 72 h post-dose. Effects of CarboCarrier® insulin were compared with those of NPH-insulin. RESULTS: CarboCarrier® insulin was safe and well-tolerated and no consistent pattern of adverse events occurred. CarboCarrier® insulin exposure (Cmax and AUC) increased proportionally with dose. The mean terminal elimination half-life ranged between 3.11 and 5.28 h. All CarboCarrier® insulin dose groups showed decreases in the mean change from baseline of plasma glucose concentrations compared with the placebo group. CONCLUSIONS: CarboCarrier® insulin is pharmacologically active showing features of insulin action in man. The elimination half-life of the molecule was clearly extended compared with endogenous insulin, indicating that conjugation to ATIII-binding pentasaccharides is a viable approach to extend the half-life of therapeutic proteins in humans. This is an important step towards validation of the CarboCarrier® technology by making use of CarboCarrier® insulin as an example.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Drug Carriers , Glycoproteins/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Insulin, Long-Acting/pharmacokinetics , Adolescent , Adult , Aged , Area Under Curve , Biological Availability , Blood Glucose/metabolism , C-Peptide/blood , Diabetes Mellitus, Type 2/drug therapy , Dose-Response Relationship, Drug , Female , Glycated Hemoglobin/metabolism , Glycoproteins/chemistry , Half-Life , Humans , Hypoglycemic Agents/chemistry , Injections, Subcutaneous , Insulin/blood , Insulin, Long-Acting/chemistry , Male , Middle Aged , Oligosaccharides/chemistry , Young Adult
12.
Eur J Clin Pharmacol ; 68(3): 249-58, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21935705

ABSTRACT

PURPOSE: The aim of our study was to evaluate the pharmacology of vorapaxar (SCH 530348), an oral PAR-1 antagonist, in healthy volunteers. METHODS AND RESULTS: In two randomized, placebo-controlled studies, subjects received either single ascending doses of vorapaxar (0.25, 1, 5, 10, 20, or 40 mg; n = 50), multiple ascending doses of vorapaxar (1, 3, or 5 mg/day for 28 days; n = 36), a loading dose (10 or 20 mg) followed by daily maintenance doses (1 mg) for 6 days (n = 12), or placebo. Single 20- and 40-mg doses of vorapaxar completely inhibited thrombin receptor activating peptide (TRAP)-induced platelet aggregation (>80% inhibition) at 1 h and sustained this level of inhibition for ≥72 h. Multiple doses yielded complete inhibition on Day 1 (5 mg/day) and Day 7 (1 and 3 mg/day). Adverse events were generally mild, transient, and unrelated to dose. CONCLUSION: Vorapaxar provided rapid and sustained dose-related inhibition of platelet aggregation without affecting bleeding or clotting times.


Subject(s)
Lactones/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Pyridines/administration & dosage , Receptor, PAR-1/antagonists & inhibitors , Adolescent , Adult , Bleeding Time , Female , Humans , Lactones/blood , Lactones/pharmacokinetics , Male , Middle Aged , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Pyridines/blood , Pyridines/pharmacokinetics , Young Adult
13.
Ned Tijdschr Geneeskd ; 154: A762, 2010.
Article in Dutch | MEDLINE | ID: mdl-20456775

ABSTRACT

A 58-year-old man, who spoke very little Dutch, had various symptoms and used several drugs including simvastatin. He was prescribed itraconazole for onychomycosis. Simvastatin was concurrently replaced with pravastatin to prevent drug interactions. However, the interaction still occurred when the pravastatin ran out, and the patient resumed taking simvastatin on his own initiative. Myalgia and muscle weakness developed after one week. The general practitioner found a strongly elevated creatine kinase level in the blood. The patient required hospitalisation for severe rhabdomyolysis. He was treated with an infusion of an ample quantity of physiological saline solution and made a full recovery. Due to the elevated risk of toxic interactions, doctors should beware of communication problems in complex patients and avoid new prescriptions not strictly required.


Subject(s)
Antifungal Agents/adverse effects , Hypolipidemic Agents/adverse effects , Itraconazole/adverse effects , Rhabdomyolysis/chemically induced , Simvastatin/adverse effects , Antifungal Agents/therapeutic use , Drug Interactions , Humans , Hypolipidemic Agents/therapeutic use , Itraconazole/therapeutic use , Male , Middle Aged , Rhabdomyolysis/therapy , Simvastatin/therapeutic use
14.
Biomed Mater Eng ; 14(4): 455-64, 2004.
Article in English | MEDLINE | ID: mdl-15472394

ABSTRACT

Glucose or lactate biosensors are very useful for monitoring metabolism. Continuous monitoring of glucose is for example very important in diabetic patients. The measurement of lactate, a marker for oxygen deficiency, is used in the intensive care unit to monitor the patients' condition. In our laboratory we have developed two types of on-line biosensors to measure in vivo glucose and lactate: a sandwich-type biosensor and, very recently, a miniaturized flow-through biosensor. These biosensors are not placed in the body itself, but are connected to implanted microdialysis or ultrafiltration probes. Both types of biosensors are based on the oxidation of substrate using glucose oxidase or lactate oxidase and electrochemical detection. In the sandwich-type sensor, the enzymes are physically immobilized between two cellulose nitrate filters, and operate with ferrocene as a mediator. In the miniaturized biosensor, with an internal volume of 10-20 nanolitres, the enzymes are immobilized on the electrode via in situ encapsulation in poly(1,3-phenylenediamine). In this review we shall explain the working of these biosensors, and describe their application in clinical monitoring and experimental research.


Subject(s)
Biosensing Techniques/instrumentation , Biotechnology/instrumentation , Blood Chemical Analysis/instrumentation , Blood Glucose/analysis , Diabetes Mellitus/blood , Diabetes Mellitus/diagnosis , Lactic Acid/blood , Biosensing Techniques/methods , Biosensing Techniques/trends , Biotechnology/methods , Biotechnology/trends , Blood Chemical Analysis/methods , Blood Chemical Analysis/trends , Equipment Design , Humans , Monitoring, Physiologic/instrumentation , Monitoring, Physiologic/methods , Monitoring, Physiologic/trends , Research/instrumentation , Research/trends , Research Design , Technology Assessment, Biomedical
15.
Curr Med Res Opin ; 20(8): 1185-95, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15324521

ABSTRACT

BACKGROUND: Ezetimibe is a lipid-lowering drug indicated for the treatment of hypercholesterolemia as co-administration with HMG-CoA reductase inhibitors (statins) or as monotherapy. The primary objectives of this study were to evaluate the pharmacodynamic effects and safety of the co-administration of ezetimibe and the new statin rosuvastatin. A secondary objective was to examine the potential for a pharmacokinetic interaction between ezetimibe and rosuvastatin. METHODS: This was a randomized, evaluator (single)-blind, placebo-controlled, parallel-group study in healthy hypercholesterolemic subjects (untreated low-density lipoprotein cholesterol [LDL-C] > or = 130 mg/dL [3.37 mmol/L]). After the outpatient screening and NCEP Step I diet stabilization periods, 40 subjects were randomized to one of the 4 following treatments: rosuvastatin 10 mg plus ezetimibe 10 mg (n = 12); rosuvastatin 10 mg plus placebo (matching ezetimibe 10 mg) (n = 12); ezetimibe 10 mg plus placebo (matching ezetimibe 10 mg) (n = 8); or placebo (2 tablets, matching ezetimibe 10 mg) (n = 8). All study treatments were administered once daily in the morning for 14 days as part of a 16-day inpatient confinement period. Fasting serum lipids were assessed pre-dose on days 1 (baseline), 7, and 14 by direct quantitative assay methods. Safety was evaluated by monitoring laboratory tests and recording adverse events. Blood samples were collected for ezetimibe and rosuvastatin pharmacokinetic evaluation prior to the first and last dose and at frequent intervals after the last dose (day 14) of study treatment. Plasma ezetimibe, total ezetimibe (ezetimibe plus ezetimibe-glucuronide) and rosuvastatin concentrations were determined by validated liquid chromatography with tandem mass spectrometric detection (LC-MS/MS) assay methods. RESULTS: All active treatments caused statistically significant (p < or = 0.02) decreases in LDL-C concentration versus placebo from baseline to day 14. The co-administration of ezetimibe and rosuvastatin caused a significantly (p < 0.01) greater reduction in LDL-C and total cholesterol than either drug alone. In this 2-week inpatient study with restricted physical activity there was no apparent effect of any treatment on high-density lipoprotein cholesterol (HDL-C) or triglycerides. The co-administration of ezetimibe and rosuvastatin caused a significantly (p < 0.01) greater percentage reduction in mean LDL-C (-61.4%) than rosuvastatin alone (-44.9%), with a mean incremental reduction of -16.4% (95%CI -26.3 to -6.53). Reported side effects were generally mild, nonspecific, and similar among treatment groups. There were no significant increases or changes in clinical laboratory tests, particularly those assessing muscle and liver function. There was no significant pharmacokinetic drug interaction between ezetimibe and rosuvastatin. CONCLUSIONS: Co-administration of ezetimibe 10 mg with rosuvastatin 10 mg daily caused a significant incremental reduction in LDL-C compared with rosuvastatin alone. Moreover, co-administering ezetimibe and rosuvastatin was well tolerated in patients with hypercholesterolemia.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Fluorobenzenes/pharmacokinetics , Hypercholesterolemia/drug therapy , Pyrimidines/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Anticholesteremic Agents/therapeutic use , Azetidines/therapeutic use , Drug Synergism , Drug Therapy, Combination , Ezetimibe , Female , Fluorobenzenes/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/blood , Male , Middle Aged , Pilot Projects , Pyrimidines/therapeutic use , Rosuvastatin Calcium , Single-Blind Method , Sulfonamides/therapeutic use , Treatment Outcome
16.
Life Sci ; 70(21): 2457-66, 2002 Apr 21.
Article in English | MEDLINE | ID: mdl-12173410

ABSTRACT

The abdominal subcutaneous interstitium is easily accessible for monitoring glucose for Diabetes Mellitus research and management. The available glucose sensing devices demand frequent blood sampling by finger pricking for calibration. Moreover, there is controversy about the exact relationship between the levels of glucose in the subcutis and blood. In the present study ultra-slow microdialysis was applied for subcutaneous fluid sampling, allowing continuous measurement of glucose in an equilibrated fluid using a nanolitre size sensor. The present method avoids in vivo calibration. During an oral glucose tolerance test glucose levels were measured simultaneously in blood, in adipose tissue and loose connective tissue layers of the abdominal subcutis in seven healthy subjects. Fasting glucose levels (mM) were 2.52 +/- 0.77 in adipose tissue and 4.67 +/- 0.17 in blood, this difference increasing to 6.40 +/- 1.57 and 11.59 +/- 1.52 at maximal glucose concentration. Moreover, the kinetics of glucose in blood and adipose tissue were different. In contrast, connective tissue glucose levels differed insignificantly (4.71 +/- 0.21 fasting and 11.70 +/- 1.96 at maximum) from those in blood and correlated well (r2 = 0.962). Ultra-slow microdialysis combined with a nanolitre glucose sensor could be of benefit to patients in intensive diabetes therapy. Frequent blood sampling for in vivo calibration can be avoided by monitoring glucose in the abdominal subcutaneous loose connective tissue, rather than in the adipose tissue.


Subject(s)
Glucose/metabolism , Skin/chemistry , Skin/metabolism , Adipose Tissue/chemistry , Adipose Tissue/metabolism , Biosensing Techniques , Blood Glucose Self-Monitoring/instrumentation , Connective Tissue/chemistry , Connective Tissue/metabolism , Glucose Tolerance Test , Humans , Insulin/blood , Microdialysis , Reference Values
17.
Clin Chim Acta ; 316(1-2): 1-10, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11750268

ABSTRACT

BACKGROUND: Tight regulation of blood glucose levels from patients suffering from diabetes mellitus can significantly reduce the complications associated with this disease. For this reason, elaborate research efforts have been devoted to the development of a glucose sensor for the continuous in vivo monitoring of glucose. Although the use of microdialysis as a sampling interface between the body and the biosensor is widely accepted, a major drawback of conventional microdialysis is the limited in vivo recovery. Here, ultraslow microdialysis is proposed in order to obtain (near) quantitative in vivo recoveries. To avoid, however, unacceptable long delay times, the need for a small and low dead volume measuring device was recognised. METHODS: A portable lightweight measuring device for continuous in vivo monitoring of glucose in subcutaneous tissue is presented. The measuring device consists of a miniaturised flow-through biosensor, connected to a microdialysis probe and a semi-vacuum pump. The biosensor is based on the amperometric detection of hydrogen peroxide after conversion of glucose by immobilised glucose oxidase. A portable potentiostat equipped with data logging is used for detection and registration. RESULTS: The device was validated for its accuracy, precision, linearity, sensitivity, selectivity and stability during ex vivo and in vivo experiments. The linearity was found to be up to 30 mmol/l with a limit of detection of 0.05 mmol/l. The precision, depending on the biosensor tested was found to be 2-4%. No contribution to the signal could be observed from several tested electroactive species. The accuracy was found to be well in accordance with the criteria set for methods of Self Monitoring of Blood Glucose for patients with diabetes mellitus. The biosensors could be used for up to 3 days in the continuous mode. In vivo monitoring of glucose in dialysate of subcutaneous sampled tissue during glucose tolerance tests in healthy volunteers demonstrated the potential of this measuring device. CONCLUSIONS: A portable lightweight measuring device is presented which can measure continuously glucose in vivo without excessive calibration steps. The performance characteristics determined justify the application of this measuring device for the in vivo monitoring of glucose in subcutaneous sampled interstitium of diabetic patients.


Subject(s)
Biosensing Techniques/instrumentation , Blood Glucose Self-Monitoring/instrumentation , Biosensing Techniques/methods , Biosensing Techniques/standards , Blood Glucose Self-Monitoring/methods , Blood Glucose Self-Monitoring/standards , Calibration , Diabetes Mellitus/blood , Dialysis , Female , Glucose Oxidase/metabolism , Glucose Tolerance Test , Humans , Hydrogen Peroxide/analysis , Miniaturization , Reproducibility of Results , Sensitivity and Specificity , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...