Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Arzneimittelforschung ; 50(9): 802-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11050696

ABSTRACT

Atenolol (CAS 29122-68-7) and chlortalidone (CAS 77-36-1) are marketed associated in a 4:1 strength ratio (100/25 and 50/12.5 mg) for the treatment of hypertension. According to EU guidelines, the bioequivalence of one dosage strength can also cover additional strengths when the pharmacokinetics of a given drug is linearly related with the dose. The kinetics of atenolol is linearly correlated with the dose and chlortalidone has linear kinetics with doses < or = 100 mg. Thus this trial carried out on the 100/25 mg strength also covers the 50/12.5 mg strength. The trial was carried out on 18 healthy volunteers (9 males and 9 females) according to a single dose, two-period, two-treatment, two-sequence study design with washout. Timed atenolol plasma concentrations and chlortalidone blood concentrations were used to assess primary pharmacokinetic parameters Cmax, tmax and AUC extrapolated to infinity by a non-compartmental model. The bioavailability of the two formulations was compared through the 90% confidence intervals (C.I.) of Cmax and AUC in accordance with operating guidelines. C.I. of chlortalidone were fully comprised in the 0.80-1.25 range. In the case of atenolol, which displayed a higher data dispersion, C.I. were comprised in the enlarged 0.70-1.43 range. Time to peak, tmax, did not show any statistically significant difference between the test and reference product with respect to both analytes. Pharmacodynamic measurements of the decrease in systolic blood pressure led to fully overlapping results with test and reference. The authors conclude that the test formulation should be considered bioequivalent with the reference with chlortalidone and in the borderline of bioequivalence with atenolol. As no safety problems were involved and pharmacodynamics led to overlapping results as between test and reference, the bioequivalence conclusion could be extended also to atenolol.


Subject(s)
Adrenergic beta-Antagonists/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Atenolol/pharmacokinetics , Benzothiadiazines , Chlorthalidone/pharmacokinetics , Sodium Chloride Symporter Inhibitors/pharmacokinetics , Adrenergic beta-Antagonists/administration & dosage , Adult , Antihypertensive Agents/administration & dosage , Area Under Curve , Atenolol/administration & dosage , Biological Availability , Chlorthalidone/administration & dosage , Diuretics , Drug Combinations , Female , Half-Life , Humans , Male , Sodium Chloride Symporter Inhibitors/administration & dosage , Therapeutic Equivalency
2.
Arzneimittelforschung ; 50(8): 688-94, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10994151

ABSTRACT

Amlodipine (CAS 88150-42-9) is a 1,4-dihydropyridine derivative, one of the most widely used drugs for the management of essential hypertension. In developing manidipine (CAS 120092-68-4), a new antihypertensive drug, amlodipine was selected as the reference comparator drug in a Phase III double blind clinical trial. However, manidipine is formulated in hard gelatin capsules, whereas amlodipine is presented as a tablet. In order to respect the double blind design of the study, it was necessary to insert the amlodipine tablet into hard gelatin capsules matching those of the new test product. This called for an amlodipine bioequivalence study on two halves of one tablet inserted into a capsule (test formulation) and two halves of one tablet ingested as such (reference formulation). The bioequivalence trial was carried out on 18 healthy volunteers (9 males and 9 females). Subjects were administered a single 10 mg dose of test and reference products according to a two-treatment, two-period, two-sequence crossover design, with a wash-out period of three weeks. Plasma concentrations of the parent compound were monitored over a period of 6 days, considering the long half-life of amlodipine. The drug was quantified with a very sensitive, robust bioassay, which was set up and validated in our laboratory. Peak concentration and area under the curve of plasma concentrations were log-transformed and analyzed to obtain 90% confidence intervals which proved to be 0.94-1.06, and thus within the acceptable bioequivalence range of 0.80-1.25. Time to peak, analyzed according to a non-parametric test, did not show any statistically significant difference between the test and reference. Both the test and reference products showed a similar and very good safety profile. The conclusion is that one amlodipine tablet broken into two halves and administered as such (reference formulation) is bioequivalent with one amlodipine tablet broken into two halves and encapsulated (test formulation).


Subject(s)
Amlodipine/administration & dosage , Antihypertensive Agents/administration & dosage , Calcium Channel Blockers/administration & dosage , Adult , Amlodipine/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Area Under Curve , Calcium Channel Blockers/pharmacokinetics , Calibration , Chromatography, Liquid , Female , Half-Life , Humans , Male , Mass Spectrometry , Therapeutic Equivalency
3.
Arzneimittelforschung ; 50(1): 43-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10683715

ABSTRACT

This paper reports the results of a pharmacokinetic study involving 24 healthy volunteers and designed to characterise the rate and extent of diclofenac absorption after the administration of a single dose of diclofenac (CAS 15307-86-5) potassium salt 50 mg in sachet (Voltfast) and tablet (Cataflam) formulations. Timed plasma concentrations of diclofenac during a 12-h-period after dosing were measured by means of HPLC with UV detection at 275 nm and a quantification limit of 10 ng/ml; the method was fully validated for pharmacokinetic purposes. These plasma concentrations were used to calculate Cmax, tmax, trapezoidal AUC0-t and AUC0-infinity and t1/2 by means of noncompartmental analysis. Cmax and tmax are the parameters expressing the rate of absorption, whereas the AUCs reflect the extent of absorption. The rate of absorption with the sachets proved to be very fast, reaching peak values at 10 min in seven subjects and at 15 min in the remaining subjects: mean time was 13.68 min, with concentrations at 5 min being 38% of Cmax. The average time to peak concentration with the tablets was 53.10 min. The extent of absorption of the sachets and tablets was similar, with AUC0-infinity values of respectively 1362 and 1214 ng.ml-1.h, and a 90% confidence interval 1.05-1.20. The highly soluble potassium salt of diclofenac was rapidly absorbed, especially in its sachet formulation, and thus appears to be an invaluable analgesic agent that is particularly useful for quick pain relief.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Diclofenac/pharmacokinetics , Administration, Oral , Adult , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Area Under Curve , Chromatography, High Pressure Liquid , Cross-Over Studies , Diclofenac/administration & dosage , Diclofenac/adverse effects , Female , Half-Life , Humans , Male , Middle Aged , Powders , Spectrophotometry, Ultraviolet , Tablets
4.
Arzneimittelforschung ; 49(12): 992-6, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10635443

ABSTRACT

This study was carried out to investigate the pharmacokinetics of zofenopril (CAS 81938-43-4) and zofenoprilat, the behaviour of the angiotensin converting enzyme (ACE) (pharmacodynamics) following the administration of zofenopril calcium at the single oral dose of 60 mg in eighteen healthy volunteers. This open label, one-way study was carried out in a single centre on 18 healthy volunteers. The volunteers received an oral single 60 mg dose of zofenopril calcium following an overnight fast. The tablet was swallowed with 250 ml of water. Fasting continued for additional 4 h after dosing and no other liquid intake was allowed from 1 h before to 2 h after administration. Plasma concentrations of zofenopril and its active metabolite zofenoprilat as well as serum ACE activity were measured before drug intake (baseline) and on timed samples over a 36 h period after dosing by LC-MS-MS, a highly sensitive, validated method for active moiety concentrations. Peak plasma concentration was reached on average at 1.19 h with zofenopril and at 1.36 h with zofenoprilat. Concentrations then decreased reaching values under or close to the limit of quantitation (1 ng.ml-1 for zofenopril, 2 ng.ml-1 for zofenoprilat) from 8 to 16 h after dosing. Complete inhibition of ACE was seen at the first blood sampling time (1 h) and lasted on average up to 9.44 h. ACE activity then slowly reactivated, but enzyme inhibition continued and was estimated to be 74% and 56% at 24 and 36 h following drug administration, respectively. From these data a complete or almost complete enzyme inhibition is expected with zofenopril given in repeated dose regimen.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Captopril/analogs & derivatives , Peptidyl-Dipeptidase A/metabolism , Adult , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Area Under Curve , Captopril/adverse effects , Captopril/pharmacokinetics , Captopril/pharmacology , Female , Half-Life , Humans , Male , Peptidyl-Dipeptidase A/blood
5.
Br J Haematol ; 95(3): 457-60, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8943884

ABSTRACT

Progenitor cell failure in the erythroid lineage is a particular problem in bone marrow failure. To provide insight into early erythopoietic development we used sensitive techniques to examine the effects of SCF, IL-3 and MIP-1 alpha on two developmentally arrested progenitor cell lines, HEL and K562. Quantitative flowcytometric analysis showed that both expressed receptors (SCF > MIP-1 alpha > IL-3). Qualitative analysis revealed HEL cells expressed more receptors than K562 cells. Clonogenic assays with sensitive haemoglobin detection showed that SCF and IL-3 did not support HEL development and reduced haemoglobin production. MIP-1 alpha reduced partially developed HEL colonies and haemoglobin in developed colonies. SCF increased development, but not haemoglobin in K562 cells, with IL-3 being more effective in both. MIP-1 alpha increased the proportion of well-developed K562 colonies but not haemoglobin. This suggests SCF, IL-3 and MIP-1 alpha all have a role to play in early erythroid cellular development, with differing actions depending on the stage of development.


Subject(s)
Erythroid Precursor Cells/cytology , Hemoglobins/metabolism , Interleukin-3/pharmacology , Macrophage Inflammatory Proteins/pharmacology , Receptors, Cytokine/metabolism , Stem Cell Factor/pharmacology , Cell Differentiation , Cell Line , Chemokine CCL4 , Colony-Forming Units Assay , Flow Cytometry , Humans
6.
Eur J Haematol ; 57(1): 72-8, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8698135

ABSTRACT

Stem cell factor (SCF) is the ligand for the dimeric c-kit tyrosine kinase receptor. Binding of SCF to c-kit is a crucial element in the developmental stimulus of late stem cells and early progenitor cells. In the erythroid lineage the SCF stimulus is important not only for proliferation and differentiation, but is also known to enhance later haemoglobin production. In an earlier report we described a rapid non-radioactive technique using the extended ester-attached labelled SCF protein itself for detecting c-kit expression in marrow and peripheral blood mononuclear populations. In the present study we have taken this a step further to analyse c-kit expression in developing erythroid cells in vitro, principally using normal donor samples. This was designed for use as a foundation for the comparison of haematological disorders. In this case we tested 4 patients with the congenital disorder of erythropoiesis, Diamond-Blackfan anaemia (DBA), finding that in all cases DBA c-kit expression was elevated over normal, in 1 case as high as 348% of the normal average. This may be indicative of the reduced state of progenitor development in these patients. These results show that the described technique is beneficial for analysis in the stem and progenitor compartment.


Subject(s)
Erythropoiesis , Fanconi Anemia/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/pharmacology , Adult , Cell Division , Colony-Forming Units Assay , Fanconi Anemia/pathology , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Recombinant Proteins , Stem Cell Factor/metabolism
7.
Br J Haematol ; 93(2): 319-25, 1996 May.
Article in English | MEDLINE | ID: mdl-8639423

ABSTRACT

Interleukin-3 (IL-3) therapy as a treatment for Diamond Blackfan anaemia (DBA) patients has been largely disappointing despite early hope it would be suitable for stimulating arrested erythropoiesis. Initial hope came from in vitro discoveries that IL-3 (+EPO) generated well-haemoglobinized BFU-E colonies in some patients, but was soon tempered by the realization that in vitro and in vivo IL-3 response did not, in the majority of cases, correlate. Nevertheless in vitro testing has been the main focus in analysing the abnormality in the stem and progenitor cell compartment in DBA. Here we report in vitro analysis of a DBA patient who responded once to IL-3 therapy, but not a second time following relapse, using short-term culture, long-term culture and c-kit analysis. Progenitor numbers before and after the first therapy were in the high normal range, but after relapse were much reduced below normal levels. Long-term cultures suggested some arrested progenitors had been reactivated into normal cycle by the first therapy, but may not have been replaced by more immature progenitors. c-kit analysis revealed increased expression in all tested cell populations. These results imply that the first IL-3 therapy reactivated some erythroid progenitors but left the progenitor pool depleted when more immature cells remained arrested.


Subject(s)
Erythroid Precursor Cells/pathology , Fanconi Anemia/pathology , Interleukin-3/therapeutic use , Proto-Oncogene Proteins c-kit/analysis , Cell Count , Cells, Cultured , Child, Preschool , Colony-Forming Units Assay , Erythropoietin/pharmacology , Fanconi Anemia/therapy , Female , Humans , Treatment Failure
8.
Br J Haematol ; 92(2): 280-6, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8602986

ABSTRACT

The congenital disorder of erythropoiesis Diamond Blackfan anaemia (DBA) exhibits a defect in the stem/progenitor cell compartment, located at the erythroid progenitor level (CFU-GEMM, BFU-E, CFU-E). Treatment of DBA with interleukin-3 (IL-3) has had limited effect, despite in vitro studies suggesting that progenitor cells were capable of responding to IL-3. Whether IL-3 is not reaching the appropriate defective target cell, the cells cannot respond, or the marrow humoral inhibitory system is overriding it, is not clear. To investigate humoral inhibitory activities we examined the response of 15 DBA bone marrows in vitro to the inhibitory chemokine macrophage inflammatory protein 1-alpha (MIP1-alpha) in the presence of the stimulatory cytokines erythropoietin, granulocyte-macrophage colony-stimulating factor, IL-3, and stem cell factor. In vitro data agreed with our previous work showing that our patients formed three statistically different groups in response to stimulatory cytokines (type I DBA erythroid colony numbers approximately normal > type II DBA > type III DBA). Addition of MIP1-alpha to cultures caused average erythroid and myeloid suppression, which sequentially increased with DBA type (type I inhibition < type II < type III). The differential level of inhibition shown by MIP1-alpha in these DBA patients lends further evidence for the presence of distinct subgroups in this disorder.


Subject(s)
Erythroid Precursor Cells/immunology , Fanconi Anemia/immunology , Monokines/pharmacology , Adolescent , Adult , Antibody Formation , Cell Division/drug effects , Chemokine CCL3 , Chemokine CCL4 , Child , Child, Preschool , Colony-Forming Units Assay , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/physiology , Erythropoietin/pharmacology , Fanconi Anemia/blood , Fanconi Anemia/classification , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/physiology , Humans , Infant , Interleukin-3/pharmacology , Macrophage Inflammatory Proteins , Male , Stem Cell Factor/pharmacology
9.
Schweiz Med Wochenschr ; 125(19): 970-4, 1995 May 13.
Article in German | MEDLINE | ID: mdl-7761808

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by T-cell activation and mucosal influx of inflammatory cells partly mediated by increased local release of cytokines and chemokines. Increased levels of activated platelets are reported in IBD. Activated platelets induce endothelial cells in vitro to secrete several cytokines and growth factors and to express adhesion molecules. This study investigates the expression of interleukin-1 (IL-1), IL-8 and granulocyte-macrophage colony-stimulating factor (GM-CSF) receptors on circulating platelets from patients with IBD and healthy controls and assesses the in vitro effect of various concentrations of IL-1 beta, IL-8 and GM-CSF on platelet activation in healthy controls. Flow cytometry was performed to quantify the percentage of platelets binding phycoerythrin (PE) labeled recombinant human IL-1 beta, IL-8 and GM-CSF. Platelet activation was assessed using fluorochrome labeled anti-GMP-140, an activation-dependent antigen. Results are expressed as percentage cytokine receptor expressing platelets (median and interquartile range IQR). Platelets from patients with IBD expressed significantly more cytokine receptors compared to healthy controls: IL-1R [8.7% (5.5-18.2) vs 3.1% (2.4-4.8), p < 0.05], IL-8R [22.5% (18.1-27.9) vs 8% (4.5-9.2), p < 0.001)], GM-CSFR [25.9% (16.1-39.2) vs 3.9% (2.7-3.9), p < 0.001]. The percentage of activated platelets was significantly increased after in vitro stimulation with IL-1 beta, IL-8 and GM-CSF. We conclude that cytokines and chemokines modulate platelet activation through specific, functional receptors which are upregulated in IBD.


Subject(s)
Inflammatory Bowel Diseases/blood , Platelet Activation/drug effects , Receptors, Cytokine/isolation & purification , Blood Platelets/chemistry , Endothelium/cytology , Endothelium/metabolism , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , In Vitro Techniques , Inflammatory Bowel Diseases/immunology , Interleukin-1/pharmacology , Interleukin-8/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/isolation & purification , Receptors, Interleukin/isolation & purification , Receptors, Interleukin-1/isolation & purification , Receptors, Interleukin-8A
10.
Exp Hematol ; 23(1): 14-20, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7527782

ABSTRACT

Stem cell factor (SCF) is a determining and crucial element in the development of early hematopoietic cells. The SCF receptor protein has been identified as the product of the protooncogene c-kit and has been detected using monoclonal antibodies (MAbs) on a broad selection of erythroid, myeloid, and lymphoid cell lines as well as on bone marrow mononuclear cells (BMMNC). SCF is known to increase both the number and size of burst-forming unit-erythroid (BFU-E) colonies in normal human BM culture in a dose-dependent fashion. A detailed study of the involvement of SCF and its receptor c-kit in normal erythropoiesis will help elucidate intrinsic irregularities of anemias such as Diamond Blackfan Anemia, an aregenerative congenital anemia. Abnormalities of this heterogeneous disorder are confined to the red cell lineage and are thought to arise through a defect at the stem/progenitor cell level. Our in vitro studies suggest that SCF therapy will influence BFU-E production in at least a portion of these patients, although in another group, SCF response is limited or absent. Additionally, further investigations have shown a possible c-kit signaling defect that clearly necessitates further c-kit characterization. To parallel this, we, therefore, attempted to study the relationship of c-kit with its ligand. This report describes a nonradioactive method for detecting SCF receptors that varies from conventional assays in that the fluorescent label conjugated to the SCF/c-kit complex is connected via an extended-ester linkage that reduces steric influence and promotes full normal structural ligand binding of the SCF to its receptor.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Bone Marrow/chemistry , Erythrocytes/chemistry , Proto-Oncogene Proteins/analysis , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Colony-Stimulating Factor/analysis , Antibodies, Monoclonal , Cell Line , Flow Cytometry , Fluorescein-5-isothiocyanate , Humans , Leukocytes, Mononuclear/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Colony-Stimulating Factor/metabolism
11.
Eur J Clin Invest ; 24(10): 656-63, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7531643

ABSTRACT

Tissue and plasma concentrations of several cytokines are increased in patients with inflammatory bowel disease (IBD). Platelets play an important role in inflammation and circulate in an activated state in patients with IBD. This study assesses the expression of IL-8 and IL-1 receptors on the surface of platelets from patients with IBD using phycoerythrin (PE)-labelled recombinant human rhIL-1 beta and rhIL-8 and flow cytometry. The percentage IL-1R expressing platelets (median and interquartile range IQR) in the IBD group was 8.7% (5.5-18.2) compared to 4.2% (2.3-6.1) in controls (P = 0.02). The percentage IL-8R expressing platelets in the IBD group was 22.5% (16.5-27.9) and 9.2% (4.3-9.6) in controls (P < 0.001). Furthermore, platelet IL-1R expression in patients with IBD was inversely related to the total daily dose of steroids (r = 0.71, P < 0.01 linear regression analysis). Finally, platelet rich plasma from healthy controls was stimulated with rhIL-1 beta and rhIL-8 and assessed for activation dependent expression of platelet aGPIIb/IIIa and CD62 (p-selectin, GMP-140). IL-1 beta and IL-8 in vitro significantly and specifically activated the platelets. The surface membrane of platelets is able to express functional IL-1R and IL-8R, the expression of which is significantly increased in IBD. Interleukin-1 beta and IL-8 modulate platelet activation in vitro indicating a target role for platelet function in inflammation.


Subject(s)
Blood Platelets/metabolism , Colitis, Ulcerative/blood , Crohn Disease/blood , Receptors, Interleukin-1/metabolism , Receptors, Interleukin/metabolism , Adolescent , Adult , Female , Humans , Interleukin-1/pharmacology , Interleukin-8/pharmacology , Male , Middle Aged , P-Selectin , Platelet Activation/drug effects , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/metabolism , Receptors, Interleukin-8A
SELECTION OF CITATIONS
SEARCH DETAIL
...