Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Bone ; 64: 204-10, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24769332

ABSTRACT

Antagonism of the calcium-sensing receptor in the parathyroid gland leads to parathyroid hormone (PTH) release. Calcilytics are a new class of molecules designed to exploit this mechanism. In order to mimic the known bone-anabolic pharmacokinetic (PK) profile of s.c. administered PTH, such molecules must trigger sharp, transient and robust release of PTH. The results of two early clinical studies with the orally-active calcilytic AXT914, a quinazolin-2ne derivative are reported. These were GCP-compliant, single and multiple dose studies of PK/PD and tolerability in healthy volunteers and postmenopausal women. The first study, examined single ascending doses (4 to 120 mg) and limited multiple doses (60 or 120 mgq.d. for 12 days) of AXT914. The second study was a randomized, double-blind, active- and placebo-controlled, 4-week repeat-dose parallel group study of healthy postmenopausal women (45 and 60 mg AXT914, placebo, 20 µg Forteo/teriparatide/PTH(1-34) fragment). AXT914 was well tolerated at all doses and reproducibly induced the desired PTH-release profiles. Yet, 4 weeks of 45 or 60 mg AXT914 did not result in the expected changes in circulating bone biomarkers seen with teriparatide. However total serum calcium levels increased above baseline in the 45 and 60 mg AXT914 treatment groups (8.0% and 10.7%, respectively), compared to that in the teriparatide and placebo groups (1.3% and 1.0%, respectively). Thus the trial was terminated after a planned interim analysis due to lack of effect on bone formation biomarkers and dose-limiting effects on serum calcium. In conclusion, AXT914 was well tolerated but the observed transient and reproducible PTH-release after repeat oral administration of AXT914 which showed an exposure profile close to that of s c. PTH, did not translate into a bone anabolic response and was associated with a persistent dose-related increase in serum calcium concentrations.


Subject(s)
Parathyroid Hormone/metabolism , Postmenopause , Quinazolinones/pharmacology , Administration, Oral , Case-Control Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Middle Aged , Placebos , Quinazolinones/administration & dosage , Quinazolinones/pharmacokinetics
2.
Bone ; 49(2): 233-41, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21514409

ABSTRACT

Parathyroid hormone (PTH), when injected daily as either the intact hormone PTH(1-84) or the active fragment PTH(1-34) (teriparatide), is an efficacious bone anabolic treatment option for osteoporosis patients. Injections lead to rapid and transient spikes in hormone exposure levels, a profile which is a prerequisite to effectively form bone. Oral antagonists of the calcium-sensing receptor (calcilytics) stimulate PTH secretion and represent thus an alternative approach to elevate hormone levels transiently. We report here on ATF936, a novel calcilytic, which triggered rapid, transient spikes in endogenous PTH levels when given orally in single doses of 10 and 30mg/kg to growing rats, and of 1mg/kg to dogs. Eight weeks daily oral application of 30mg/kg of ATF936 to aged female rats induced in the proximal tibia metaphysis increases in bone mineral density, cancellous bone volume and cortical and trabecular thickness as evaluated by computed tomography. In healthy humans, single oral doses of ATF936 produced peak PTH levels in plasma after a median time of 1h and levels returned to normal at 24-h post-dose. The average maximum PTH concentration increase from baseline was 1.9, 3.6, and 6.0-fold at doses of 40, 70, and 140mg. ATF936 was well tolerated. The sharp, transient increase in PTH levels produced by the oral calcilytic ATF936 was comparable to the PTH profile observed after subcutaneous administration of teriparatide. In conclusion, ATF936 might hold potential as an oral bone-forming osteoporosis therapy.


Subject(s)
Anabolic Agents/pharmacology , Bone Density/drug effects , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Quinazolinones/pharmacology , Receptors, Calcium-Sensing/antagonists & inhibitors , Adult , Anabolic Agents/pharmacokinetics , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Calcium/metabolism , Dogs , Female , Humans , Male , Molecular Structure , Quinazolinones/pharmacokinetics , Rats , Rats, Wistar
3.
Drug Metab Dispos ; 38(5): 808-16, 2010 May.
Article in English | MEDLINE | ID: mdl-20097723

ABSTRACT

Deferasirox (ICL670) is a novel once-daily, orally administered iron chelator to treat chronic iron overload in patients with transfusion-dependent anemias. Absorption, distribution, metabolism, and excretion of [14C]deferasirox at pharmacokinetic steady state was investigated in five adult beta-thalassemic patients. Deferasirox (1000 mg) was given orally once daily for 6 days to achieve steady state. On day 7, patients received a single oral 1000-mg dose (approximately 20 mg/kg) of [14C]deferasirox (2.5 MBq). Blood, plasma, feces, and urine samples collected over 7 days were analyzed for radioactivity, deferasirox, its iron complex Fe-[deferasirox]2, and metabolites. Deferasirox was well absorbed. Deferasirox and its iron complex accounted for 87 and 10%, respectively, of the radioactivity in plasma (area under the curve at steady state). Excretion occurred largely in the feces (84% of dose), and 60% of the radioactivity in the feces was identified as deferasirox. Apparently unchanged deferasirox in feces was partly attributable to incomplete intestinal absorption and partly to hepatobiliary elimination of deferasirox (including first-pass elimination) and of its glucuronide. Renal excretion was only 8% of the dose and included mainly the glucuronide M6. Oxidative metabolism by cytochrome 450 enzymes to M1 [5-hydroxy (OH) deferasirox, presumably by CYP1A] and M4 (5'-OH deferasirox, by CYP2D6) was minor (6 and 2% of the dose, respectively). Direct and indirect evidence indicates that the main pathway of deferasirox metabolism is via glucuronidation to metabolites M3 (acyl glucuronide) and M6 (2-O-glucuronide).


Subject(s)
Benzoates/metabolism , Benzoates/pharmacokinetics , Iron Overload/drug therapy , Transfusion Reaction , Triazoles/metabolism , Triazoles/pharmacokinetics , beta-Thalassemia/therapy , Adult , Animals , Area Under Curve , Arylsulfatases/metabolism , Benzoates/adverse effects , Benzoates/therapeutic use , Cells, Cultured , Deferasirox , Feces/chemistry , Female , Glucuronidase/metabolism , Glucuronides/analysis , Glucuronides/metabolism , Hepatocytes/enzymology , Humans , Hydroxylation , Iron Chelating Agents/adverse effects , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/therapeutic use , Male , Molecular Structure , Rats , Spectrometry, Mass, Electrospray Ionization , Sulfuric Acid Esters/analysis , Sulfuric Acid Esters/metabolism , Triazoles/adverse effects , Triazoles/therapeutic use , Young Adult
4.
Drug Metab Dispos ; 36(12): 2523-38, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18775980

ABSTRACT

Deferasirox (Exjade, ICL670, CGP72670) is an iron-chelating drug for p.o. treatment of transfusional iron overload in patients with beta-thalassemia or sickle cell disease. The pharmacokinetics and disposition of deferasirox were investigated in rats. The animals received single intravenous (10 mg/kg) or p.o. (10 or 100 mg/kg) doses of 14C-radiolabeled deferasirox. Biological samples were analyzed for radioactivity (liquid scintillation counting, quantitative whole-body autoradioluminography), for deferasirox and its iron complex [high-performance liquid chromatography (HPLC)/UV], and for metabolites (HPLC with radiodetection, liquid chromatography/mass spectrometry, 1H and 13C NMR, and two-dimensional NMR techniques). At least 75% of p.o.-dosed deferasirox was absorbed. The p.o. bioavailability was 26% at the 10 mg/kg dose and showed an overproportional increase at the 100 mg/kg dose, probably because of saturation of elimination processes. Deferasirox-related radioactivity was distributed mainly to blood, excretory organs, and gastrointestinal tract. Enterohepatic recirculation of deferasirox was observed. No retention occurred in any tissue. The placental barrier was passed to a low extent. Approximately 3% of the dose was transferred into the breast milk. Excretion of deferasirox and metabolites was rapid and complete within 7 days. Key clearance processes were hepatic metabolism and biliary elimination via multidrug resistance protein 2. Deferasirox, iron complex, and metabolites were excreted largely via bile and feces (total > or = 90%). Metabolism included glucuronidation at the carboxylate group (acyl glucuronide M3) and at phenolic hydroxy groups, as well as, to a lower degree, cytochrome P450-catalyzed hydroxylations. Two hydroxylated metabolites (M1 and M2) were administered to rats and were shown not to contribute substantially to iron elimination in vivo.


Subject(s)
Benzoates/metabolism , Benzoates/pharmacokinetics , Iron/metabolism , Triazoles/metabolism , Triazoles/pharmacokinetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Animals, Suckling/metabolism , Benzoates/administration & dosage , Bile/chemistry , Blood Chemical Analysis , Deferasirox , Enterohepatic Circulation , Feces/chemistry , Female , Fetus/metabolism , Iron/analysis , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacology , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Milk, Human/chemistry , Milk, Human/metabolism , Models, Biological , Molecular Structure , Placenta/metabolism , Pregnancy , Rats , Rats, Long-Evans , Rats, Transgenic , Rats, Wistar , Tissue Distribution , Triazoles/administration & dosage , Urine/chemistry
5.
J Chromatogr A ; 1133(1-2): 184-94, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16970958

ABSTRACT

Sensitive radioactivity detection following high performance liquid chromatography (HPLC) separation remains a challenge in many drug metabolism studies with radiolabeled compounds. In this work, solid scintillation counting (SSC) after fraction collection into 96-well plates was evaluated as an off-line radioactivity detection method, in comparison with conventional liquid scintillation counting (LSC). The impact of counting time and biological matrix on the quantification of radiolabeled metabolites and parent drug in samples from animal and human absorption, distribution, metabolism and excretion (ADME) studies was investigated. Three different approaches were used to test whether reliable quantification by off-line SSC detection, which requires an approximately constant counting yield during the entire chromatographic run, can be realized: (i) the measurement of radioactivity-spiked biological blank samples without HPLC separation as an extreme case of biological background, (ii) the measurement of radioactivity-spiked HPLC fractions of biological blank samples and (iii) the comparison of radiochromatograms obtained by off-line SSC and LSC of real samples from ADME studies with radiolabeled compounds. Situations in which variations in SSC yield during an HPLC run are likely to lead to significant errors in quantitation were identified and are discussed. However, examples from a number of animal or human ADME studies showed that in the majority of cases off-line SSC provides very similar quantitative data, compared with the reference method of off-line LSC radioactivity detection. Approaches for validation of the off-line SSC approach in critical cases are discussed. The main advantages of off-line SSC, compared with off-line LSC, are lower detection limits and a substantially higher throughput. Several applications of off-line SSC detection in ADME studies are shown.


Subject(s)
Chromatography, High Pressure Liquid/methods , Pharmaceutical Preparations/analysis , Radioisotopes/analysis , Scintillation Counting/methods , Animals , Pharmaceutical Preparations/chemistry , Radioisotopes/chemistry , Rats , Reproducibility of Results
6.
Electrophoresis ; 23(9): 1255-62, 2002 May.
Article in English | MEDLINE | ID: mdl-12007124

ABSTRACT

Highly efficient capillary electrochromatographic separations of cardiac glycosides and other steroids are presented. Employing butyl-derivatized silica particles as stationary phase resulted in a nearly three times faster electroosmotic flow (EOF) compared to capillary electrochromatography (CEC) with octadecyl silica particles. On-column focusing with a preconcentration factor of 180 was performed and separation efficiencies of up to 240,000 plates per meter were obtained. Using label-free standard UV absorbance, detection limits of 10-80 nM were reached for all steroids tested. For screening of cardiac glycosides, e.g., digoxin and digitoxin in mixtures of steroids, CEC was combined with immunoaffinity extraction using immobilized polyclonal anti-digoxigenin antibodies and F(ab) fragments. Simply adding small amounts of antibody carrying particles to the samples and comparing chromatograms before and after antibody addition allowed screening for high affinity antigens in mixtures with moderate numbers of compounds. Under conditions of competing antigens, affinity fingerprints of immobilized anti-digoxigenin and anti-digitoxin antibodies were obtained, reflecting the cross-reactivity of eleven steroids. The method provides high selectivity due to the combination of bioaffinity interaction with highly efficient CEC separation and UV detection at several wavelengths in parallel. This selectivity was exploited for the detection of four cardiac glycosides in submicromolar concentrations in an untreated urine sample.


Subject(s)
Cardiac Glycosides/isolation & purification , Electrophoresis, Capillary/methods , Steroids/isolation & purification , Antibodies , Cardiac Glycosides/immunology , Chromatography, Affinity , Chromatography, High Pressure Liquid , Digitoxin/immunology , Digitoxin/isolation & purification , Digoxigenin/immunology , Digoxigenin/isolation & purification , Digoxin/immunology , Digoxin/isolation & purification , Immunoglobulin Fab Fragments , Immunosorbent Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...