Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Thromb Haemost ; 14(7): 1410-9, 2016 07.
Article de Anglais | MEDLINE | ID: mdl-27371116

RÉSUMÉ

UNLABELLED: Essentials ADAMTS-13-deficiency is a cause of thrombotic thrombocytopenic purpura (TTP). Preclinical safety of recombinant human ADAMTS-13 (BAX930) was shown in animal models. Preclinical efficacy of BAX930 was shown in a mouse model of TTP. BAX930 showed advantageous efficacy over fresh frozen plasma, the current standard of care. Click to hear Dr Cataland and Prof. Lämmle present a seminar on Thrombotic Thrombocytopenic Purpura (TTP): new Insights in Pathogenesis and Treatment Modalities. SUMMARY: Background Thrombotic thrombocytopenic purpura (TTP) is a rare blood disorder characterized by microthrombosis in small blood vessels of the body, resulting in a low platelet count. Baxalta has developed a new recombinant ADAMTS-13 (rADAMTS-13) product (BAX930) for on-demand and prophylactic treatment of patients with hereditary TTP (hTTP). Objectives To evaluate the pharmacokinetics, efficacy and safety of BAX930 in different species, by use of an extensive preclinical program. Methods The prophylactic and therapeutic efficacies of BAX930 were tested in a previously established TTP mouse model. Pharmacokinetics were evaluated after single intravenous bolus injection in mice and rats, and after repeated dosing in cynomolgus monkeys. Toxicity was assessed in rats and monkeys, safety pharmacology in monkeys, and local tolerance in rabbits. Results BAX930 was shown to be efficacious, as demonstrated by a stabilized platelet count in ADAMTS-13 knockout mice that were thrombocytopenic when treated. Prophylactic efficacy was dose-dependent and comparable with that achieved by treatment with fresh frozen plasma, the mainstay of hTTP treatment. Therapeutic efficacy was treatment interval-dependent. Safety pharmacology evaluation did not show any deleterious effects of BAX930 on cardiovascular and respiratory functions in monkeys. The compound's pharmacokinetics were similar and dose-proportional in mice, rats, and monkeys. BAX930 was well tolerated in rats, monkeys, and rabbits, even at the highest doses tested. Conclusions These results demonstrate that BAX930 has a favorable preclinical profile, and support the clinical development of rADAMTS-13 for the treatment of hTTP.


Sujet(s)
Protéine ADAMTS13/pharmacologie , Purpura thrombotique thrombocytopénique/traitement médicamenteux , Protéine ADAMTS13/génétique , Animaux , Aire sous la courbe , Plaquettes/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Femelle , Humains , Macaca fascicularis , Mâle , Souris , Plasma sanguin/métabolisme , Numération des plaquettes , Purpura thrombotique thrombocytopénique/sang , Lapins , Rats , Protéines recombinantes/pharmacologie , Spécificité d'espèce , Thrombose/sang , Résultat thérapeutique
2.
Haemophilia ; 21(1): 58-63, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25459309

RÉSUMÉ

Prophylaxis prevents joint and other bleeding episodes in patients with haemophilia A. Development of new factor concentrates with longer circulating half-lives may encourage patients to start, continue or resume prophylaxis. The aim of this study was to compare the pharmacodynamic effect of a PEGylated full-length recombinant factor VIII (rFVIII) concentrate with that of an unmodified rFVIII concentrate with respect to the duration of prophylactic efficacy in a murine model of haemophilic joint bleeding. Mice were pretreated with BAX 855 or unmodified rFVIII at specified times before right knee puncture to induce haemarthrosis; left knee joints served as controls. Joint bleeding was evaluated using a combination of visual and histological assessments. Administration of a single dose of unmodified rFVIII before joint puncture prevented haemarthrosis in mice up to 24 h, whereas pretreatment with BAX 855 protected the joint from bleeding up to 48 h. This pharmacodynamic study showed prolonged efficacy of BAX 855 compared to ADVATE in a haemophilia A mouse joint bleeding model. This finding supports the possibility of using BAX 855 to increase FVIII trough levels and/or extend the dosing interval in patients with haemophilia A on prophylaxis, which may potentially improve prophylactic efficacy and long-term adherence.


Sujet(s)
Facteur VIII/administration et posologie , Hémophilie A/traitement médicamenteux , Animaux , Modèles animaux de maladie humaine , Hémorragie/prévention et contrôle , Humains , Souris , Protéines recombinantes/administration et posologie
4.
Hamostaseologie ; 32 Suppl 1: S29-38, 2012.
Article de Anglais | MEDLINE | ID: mdl-22961422

RÉSUMÉ

A longer acting recombinant FVIII is expected to serve patients' demand for a more convenient prophylactic therapy. We have developed BAX 855, a PEGylated form of Baxter's rFVIII product ADVATE™ based on the ADVATE™ manufacturing process. The conjugation process for preparing BAX 855 uses a novel PEG reagent. The production process was adjusted to yield a rFVIII conjugate with a low PEGylation degree of about 2 moles PEG per FVIII molecule. This optimised modification degree resulted in an improved PK profile for rFVIII without compromising its specific activity. PEGylation sites were identified by employing various HPLC- and MS-based methods. These studies not only indicated that about 60% of the PEG chains are localised to the B-domain, which is cleaved off upon physiological activation during the coagulation process, but also demonstrated an excellent lot to lot consistency with regard to PEGylation site distribution. Detailed biochemical characterization further showed that PEGylated FVIII retained all the physiological functions of the FVIII molecule with the exception of binding to the LRP clearance receptor which was reduced for BAX 855 compared to ADVATE™. This might provide an explanation for the prolonged circulation time of BAX 855 as reduced receptor binding might slow-down clearance. Preclinical studies showed improved pharmacokinetic behaviour and clinically relevant prolonged efficacy compared to ADVATE™ without any signs of toxicity or elevated immunogenicity. The comprehensive preclinical data package formed the basis for approval of the phase 1 clinical study by European authorities which started in 2011.


Sujet(s)
Conception de médicament , Facteur VIII/administration et posologie , Facteur VIII/composition chimique , Hémophilie A/traitement médicamenteux , Liposomes/composition chimique , Polyéthylène glycols/composition chimique , Relation dose-effet des médicaments , Stabilité de médicament , Facteur VIII/pharmacocinétique , Période , Hémophilie A/métabolisme , Humains , Protéines recombinantes/administration et posologie , Protéines recombinantes/composition chimique , Protéines recombinantes/pharmacocinétique
5.
Vox Sang ; 100(3): 285-97, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-20946535

RÉSUMÉ

BACKGROUND AND OBJECTIVES: A human plasma-derived butyrylcholinesterase preparation manufactured on the industrial scale is described. MATERIAL AND METHODS: The human butyrylcholinesterase (hBChE) product was extensively investigated for its purity using immunological and electrophoretic methods and characterized by thorough glycoproteomic approaches. A comprehensive preclinical testing programme addressing safety and pharmacokinetic parameters supplemented the biochemical characterization. RESULTS: The high-purity hBChE preparation is tetrameric and has high specific activity and molecular integrity of the protein backbone. Acute toxicity studies and in vivo thrombogenicity studies provided evidence of a sufficient safety margin for use in humans. CONCLUSION: Extensive preclinical safety and pharmacokinetic testing confirmed that this hBChE preparation can be used for further efficacy testing as a bioscavenger for toxic organophosphate compounds in appropriate animal models and ultimately in humans.


Sujet(s)
Butyrylcholine esterase/isolement et purification , Industrie pharmaceutique/méthodes , Butyrylcholine esterase/pharmacocinétique , Butyrylcholine esterase/toxicité , Humains , Test de matériaux , Organophosphates , Pharmacocinétique , Contrôle de qualité , Virus
7.
Haemophilia ; 16(102): 25-34, 2010 May.
Article de Anglais | MEDLINE | ID: mdl-20536983

RÉSUMÉ

The development of inhibitory antibodies against factor VIII (FVIII) is the major complication in patients with haemophilia A who are treated with FVIII products. Memory B cells play an essential role in maintaining established antibody responses. Upon re-exposure to the same antigen, they are rapidly re-stimulated to proliferate and differentiate into antibody-secreting plasma cells (ASC) that secrete high-affinity antibodies. It is, therefore, reasonable to believe that memory B cells have to be eradicated or inactivated for immune tolerance induction therapy to be successful in patients with haemophilia A and FVIII inhibitors. The aim of our studies was the development of strategies to prevent FVIII-specific memory B cells from becoming re-stimulated. We established a 6-day in vitro culture system that enabled us to study the regulation of FVIII-specific murine memory-B-cell re-stimulation. We tested the impact of the blockade of co-stimulatory interactions, of different concentrations of FVIII and of ligands for toll-like receptors (TLR). The blockade of B7-CD28 and CD40-CD40 ligand interactions prevented FVIII-specific murine memory B cells from becoming re-stimulated by FVIII in vitro and in vivo. Furthermore, high concentrations of FVIII blocked re-stimulation of FVIII-specific murine memory B cells. Triggering of TLR7 amplified re-stimulation by low concentrations of FVIII and prevented blockade by high concentrations of FVIII. We conclude that we defined modulators that either amplify or inhibit the re-stimulation of FVIII-specific murine memory B cells. Currently, we are investigating whether the same modulators operate in patients with haemophilia A and FVIII inhibitors.


Sujet(s)
Lymphocytes B/immunologie , Facteur VIII/immunologie , Hémophilie A/immunologie , Mémoire immunologique/immunologie , Adolescent , Adulte , Animaux , Anticorps/immunologie , Antigènes CD/immunologie , Lymphocytes B/cytologie , Ligand de CD40/immunologie , Différenciation cellulaire , Enfant , Facteur VIII/administration et posologie , Facteur VIII/antagonistes et inhibiteurs , Hémophilie A/thérapie , Humains , Activation des lymphocytes/immunologie , Souris , Rate/cytologie , Rate/immunologie , Jeune adulte
8.
Pharmazie ; 65(4): 267-73, 2010 Apr.
Article de Anglais | MEDLINE | ID: mdl-20432623

RÉSUMÉ

Since passive immunization with serum-derived immunoglobulins (intravenous immunoglobulins) showed several positive effects in some patients with Alzheimer's disease (AD), intravenous immunoglobulins (IVIG) are discussed as a possible treatment option. IVIG, an antibody product derived from human plasma, contains natural antibodies against amyloid beta(Abeta) peptide. Until now it is not known, how IVIG interferes with pathogenesis in AD, but several proposed mechanisms are in discussion. Receptor types which are involved in transport processes at the BBB are LRP, RAGE and hFcRn. We were looking for an in vitro BBB model expressing these receptors and studied the alteration of transport of Abeta peptides across this model under the influence of immunoglobulins. Cell line ECV304 was found to be suitable for our experiments. We found evidence for involvement of an improved clearance of Abeta across the BBB as well as a decreased Abeta influx from blood to the brain probably following complex formation of immunoglobulins with free Abeta in the periphery. Furthermore, we were able to confirm the activity of IVIG preparations which acted the same way but showed slightly less efficacy in comparison to monoclonal anti-Abeta antibodies. Based on these results we suggest multiple mechanisms responsible for the efficacy of immunotherapy in Alzheimer's disease.


Sujet(s)
Peptides bêta-amyloïdes/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Immunoglobulines/sang , Immunoglobulines/pharmacologie , Algorithmes , Peptides bêta-amyloïdes/analyse , Animaux , Transport biologique actif , Technique de Western , Lignée cellulaire , Électrophorèse sur gel de polyacrylamide , Technique d'immunofluorescence , Produits terminaux de glycation avancée/métabolisme , Humains , Sérums immuns/composition chimique , Perméabilité , Rats , Récepteur Fc/effets des médicaments et des substances chimiques
9.
Lab Anim ; 44(3): 211-7, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-20507877

RÉSUMÉ

The tail cut bleeding model (CUT) is routinely used in factor VIII-deficient mice to assess pharmacodynamic effects of therapeutic strategies for haemophilia A. Results from this model are highly variable, many modifications to the model are reported and at times the animals' wellbeing may be compromised by recording survival as an endpoint. We therefore investigated if the ferric chloride carotid occlusion model (COM) used for thrombosis research can be applied to enhance data quality and animal welfare in haemophilia A research. Relative dose effects and relative dose variations were calculated for the CUT and COM. The requisite sample sizes were estimated and the importance of survival rates to assess rebleeds during recovery was evaluated by correlating initial blood loss to mortality. Relative dose effects increased with higher doses in both models. The COM was more sensitive at lower doses than the CUT, had up to 82% less variation across doses and clearly showed superior accuracy. Only 5% of the sample size required for the CUT would be needed to establish non-inferiority between a specific therapeutic dose in haemophilia A mice and healthy wild-type animals. A strong statistically significant correlation was found between initial blood loss and mortality within 24 h. Our findings clearly suggest that the COM is a valid tool for assessing haemophilia A treatment in vivo. The highly reproducible data means that significantly fewer animals are required and a more humane endpoint can be used by directly assessing clot stability instead of survival rate.


Sujet(s)
Alternatives à l'utilisation d'animaux , Bien-être animal , Coagulants/pharmacologie , Hémophilie A/traitement médicamenteux , Plan de recherche , Animaux , Artériopathies oblitérantes/induit chimiquement , Artériopathies oblitérantes/traitement médicamenteux , Artériopathies oblitérantes/anatomopathologie , Artériopathies carotidiennes/induit chimiquement , Artériopathies carotidiennes/traitement médicamenteux , Artériopathies carotidiennes/anatomopathologie , Chlorures/toxicité , Modèles animaux de maladie humaine , Femelle , Composés du fer III/toxicité , Hémophilie A/induit chimiquement , Mâle , Souris , Lignées consanguines de souris , Souris knockout , Débit sanguin régional/effets des médicaments et des substances chimiques
10.
Hamostaseologie ; 29 Suppl 1: S32-8, 2009 Oct.
Article de Anglais | MEDLINE | ID: mdl-19763356

RÉSUMÉ

Baxter has developed a recombinant therapy for treating von Willebrand's disease. Recombinant VWF is co-expressed with the rFVIII in CHO cells used to produce the rFVIII product Advate. This rVWF is used as a drug component for a rVWF-rFVIII complex drug product. CHO cells produce partially processed and partially un-processed rVWF still containing the pro-peptide. In order to make a consistent preparation containing mature and processed rVWF only rVWF is exposed to recombinant furin to remove the pro-peptide. Recombinant VWF and furin are produced under serum- and protein-free conditions. It is highly purified by a series of chromatographic steps and formulated in a protein-free buffer and has a homogeneous multimer distribution. The specific activity is higher in rVWF than in commercial plasma-derived VWF-FVIII complex products. SDS agarose electrophoretic analysis shows the presence of ultra-high molecular weight multimers. The FVIII-binding capacity and affinity of rVWF to FVIII is comparable to VWF in plasma. Carbohydrate analysis shows an intact glycosylation pattern. Recombinant VWF binds to collagen and promotes platelet adhesion under shear stress. It stabilizes endogenous FVIII in VWF-deficient knock-out mice as seen by a secondary rise in murine FVIII.


Sujet(s)
Protéines recombinantes/composition chimique , Facteur de von Willebrand/composition chimique , Albumines/composition chimique , Animaux , Aire sous la courbe , Cellules CHO , Cricetinae , Cricetulus , Modèles animaux de maladie humaine , Chiens , Facteur VIII/métabolisme , Période , Humains , Souris , Souris knockout , Plasma sanguin/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/isolement et purification , Protéines recombinantes/métabolisme , Protéines recombinantes/pharmacocinétique , Suidae , Maladies de von Willebrand/traitement médicamenteux , Facteur de von Willebrand/génétique , Facteur de von Willebrand/isolement et purification , Facteur de von Willebrand/métabolisme , Facteur de von Willebrand/pharmacocinétique
11.
J Thromb Haemost ; 7(7): 1134-42, 2009 Jul.
Article de Anglais | MEDLINE | ID: mdl-19422458

RÉSUMÉ

BACKGROUND: von Willebrand factor (VWF) is composed of a series of multimers, the sizes of which are regulated by the plasma metalloprotease ADAMTS13. OBJECTIVE: Proteolysis of human recombinant VWF (rVWF) by ADAMTS13 present in the plasma of different species typically used as preclinical animal models was investigated to evaluate the efficacy and safety of rVWF. METHODS: Degradation of rVWF was studied in vitro under moderate denaturing conditions and was monitored by multimer analysis, residual collagen binding, and immunoblot analysis. In vivo cleavage was determined by administration of rVWF to cynomolgus monkeys, rabbits and VWF-deficient mice and subsequent analysis of plasma samples by immunoblot. Plasma ADAMTS13 levels were determined with a synthetic human VWF peptide (FRETS-VWF73). RESULTS: From the animals tested, only rabbit plasma was as efficient as human plasma in proteolysing rVWF in vitro. Mouse plasma virtually failed to cleave rVWF. Administration of human rVWF resulted in ADAMTS13-specific cleavage products in rabbits and, to a lesser extent, in cynomolgus monkeys at various doses of rVWF. Virtually no cleavage occurred in mice. ADAMTS13 activity levels in rabbit and monkey plasma were similar to those in human plasma and were not significantly altered upon infusion of rVWF up to very high doses, indicating that rVWF did not lead to an exhaustion of endogenous ADAMTS13 in both species. CONCLUSIONS: The differences in susceptibility to cleavage of rVWF by different species need to be considered when interpreting the physiology of human rVWF from results of tests in animal models.


Sujet(s)
Protéines ADAM/métabolisme , Facteur de von Willebrand/métabolisme , Protéines ADAM/sang , Protéine ADAMTS13 , Animaux , Technique de Western , Humains , Hydrolyse , Macaca fascicularis , Souris , Lapins , Protéines recombinantes/métabolisme , Spécificité d'espèce
12.
J Thromb Haemost ; 5(4): 670-9, 2007 Apr.
Article de Anglais | MEDLINE | ID: mdl-17403201

RÉSUMÉ

Murine blood coagulation factors and function are quite similar to those of humans. Because of this similarity and the adaptability of mice to genetic manipulation, murine coagulation factors and inhibitors have been extensively studied. These studies have provided significant insights into human hemostasis. They have also provided useful experimental models for evaluation of the pathophysiology and treatment of thrombosis. This review contains recommendations for obtaining, processing and assaying mouse blood hemostatic components, and it summarizes the extensive literature on murine coagulation factor structure and function, assays and genetic alteration. It is intended to be a convenient reference source for investigators of hemostasis and thrombosis.


Sujet(s)
Modèles animaux de maladie humaine , Animaux , Coagulation sanguine , Fibrinogène/génétique , Hémostase/génétique , Humains , Souris , Modèles biologiques , Modèles génétiques , Temps partiel de thromboplastine , Temps de prothrombine , Thrombose/génétique
13.
Vox Sang ; 92(1): 42-55, 2007 Jan.
Article de Anglais | MEDLINE | ID: mdl-17181590

RÉSUMÉ

BACKGROUND AND OBJECTIVES: The ultimate goal was to generate an industrial-scale process suitable to produce a high-yield, safe and stable immunoglobulin G (IgG) preparation for intravenous administration, which is ready to use for customer convenience. This new liquid 10% IgG preparation (IGIV 10%) was compared to Gammagard SD, a licenced lyophilized immunoglobulin in biochemical and preclinical testing. MATERIALS AND METHODS: The new process, which includes three dedicated virus clearance steps, is a streamlined combination of the currently applied and well-established manufacturing procedures. The biochemical characterization is done by standard methods focusing on purity, integrity and functionality of the preparation. Efficacy is demonstrated in vivo by mouse protection testing and in vitro by opsonization and protein A affinity chromatography. Pharmacokinetics in rats is evaluated after a single intravenous dose. The anaphylactoid potential is determined in rats and in guinea pigs, while thrombogenicity is assessed in a rabbit model. The influence of the products on vital functions is tested on dogs, while acute toxicity studies are carried out on mice and rats. RESULTS: The biochemical characterization data demonstrate the high purity of monomeric IgG in the product. The mouse protection test showed that the protective activity against systemic bacterial infections of IGIV 10% is at least as good as the reference Gammagard SD. This result is supported by the broad spectrum of antibodies in high titres against bacteria and viruses and the high functional integrity of the IgG molecule (> or = 90% functionally intact IgG) in IGIV 10%. The opsonic activity of all IGIV 10% lots is similar to the one of the reference Gammagard SD. In safety and thrombogenicity studies, no adverse effects of IGIV 10% were observed. Pharmacokinetic studies showed no statistically significant differences between the two products. In the acute toxicity animal studies, IGIV 10% compared favourably to the reference Gammagard SD. CONCLUSIONS: The new manufacturing process enables the production of a highly purified IgG preparation for intravenous administration. The product has an IgG subclass distribution similar to plasma and contains a broad spectrum of functionally intact antibodies. Preclinical studies demonstrate that the liquid IGIV 10% combines excellent qualities of efficacy, safety and tolerability.


Sujet(s)
Décontamination/méthodes , Désinfection/méthodes , Immunoglobulines par voie veineuse/isolement et purification , Facteurs immunologiques/isolement et purification , Préparations pharmaceutiques/isolement et purification , Animaux , Chiens , Évaluation préclinique de médicament , Effets secondaires indésirables des médicaments , Humains , Immunoglobulines par voie veineuse/composition chimique , Immunoglobulines par voie veineuse/pharmacocinétique , Facteurs immunologiques/composition chimique , Facteurs immunologiques/pharmacocinétique , Souris , Préparations pharmaceutiques/sang , Préparations pharmaceutiques/composition chimique , Lapins , Rats , Résultat thérapeutique
14.
Arzneimittelforschung ; 50(11): 987-94, 2000 Nov.
Article de Anglais | MEDLINE | ID: mdl-11148865

RÉSUMÉ

alpha 1-Acid glycoprotein (AAG), a highly negatively charged glycoprotein, well known for its capillary stabilizing effect, was tested in rat models of acute edematous pancreatitis, acute hemorrhagic-necrotizing pancreatitis, and acute respiratory distress syndrome (ARDS). In cerulein-elicited edematous pancreatitis AAG improved histological alterations at 200 mg/kg i.v. and plasma amylase activity at 1800 or 4200 mg/kg i.v. All other parameters (edema, plasma lipase) were not affected in a biologically relevant manner. In glycodeoxycholic acid-induced hemorrhagic-necrotizing pancreatitis AAG was without effect on parameters measured (plasma amylase, plasma lipase activity, histological scores) at 1800 or 4200 mg/kg i.v. At the extremely high dose of 1500 mg/kg i.v. plasma amylase and lipase levels were decreased. In lipopolysaccharide-mediated ARDS, AAG was tested at 50, 200 or 600 mg/kg i.v. AAG, but also the placebo formulation decreased the myeloperoxidase content in the bronchoalveolar lavage fluid. Histological alterations were improved by AAG, however, not by the placebo formulation. Lung water content was not significantly influenced by AAG, whereas Evans blue extravasation was significantly diminished by all three doses of AAG. It is concluded that the edematous pancreatitis is the first in vivo condition with increased extravascular fluid accumulation, in which AAG is not effective. Based on data presented here and literature data, there is evidence for a beneficial effect of AAG in acute lung injury.


Sujet(s)
Maladies pulmonaires/traitement médicamenteux , Orosomucoïde/usage thérapeutique , Pancréatite/traitement médicamenteux , Maladie aigüe , Animaux , Liquide de lavage bronchoalvéolaire/cytologie , Céruléine , Oedème/induit chimiquement , Oedème/prévention et contrôle , Acide glycodésoxycholique , Hémorragie/induit chimiquement , Hémorragie/anatomopathologie , Lipopolysaccharides , Maladies pulmonaires/induit chimiquement , Mâle , Pancréatite/induit chimiquement , Rats , Rat Sprague-Dawley , Maladies de l'appareil respiratoire/induit chimiquement , Maladies de l'appareil respiratoire/traitement médicamenteux
16.
Wien Klin Wochenschr ; 111(5): 192-8, 1999 Mar 12.
Article de Allemand | MEDLINE | ID: mdl-10226349

RÉSUMÉ

The molecular properties of alpha 1-acid glycoprotein are briefly discussed. This molecule has been shown in in vitro experiments to have both a stabilizing effect on vascular permeability and antiinflammatory properties. We were able to demonstrate these two effects in vivo in guinea pigs (skin, Evan's Blue extravasation) and in rats (paw, carrageenan induced inflammation). Further experiments were performed in rats relating to possible therapeutic indications for alpha 1-acid glycoprotein: (1) inhibitory effect on brain edema formation after experimental stroke, (2) therapeutic effect in the puromycin aminonucleoside-induced minimal change nephrosis, (3) improvement of vital parameters in hemorrhagic-hypovolemic shock, (4) increase in survival rate in septic peritonitis, and (5) promising effects in burn-induced remote lung injury. The high content of sialic acid and the high negative charge of alpha 1-acid glycoprotein are believed to be major contributors to its stabilizing effect on vascular permeability. The protein is bound to the glycocalyx of the endothelial cells (and presumably to structures of the glomerular basement membrane), thereby hindering the passage of other polyanionic molecules through the vascular wall. The antiinflammatory/immunomodulatory effect of alpha 1-acid glycoprotein appears mainly due to suppression of polymorphonuclear neutrophils. This action is dependent on the glycan part of the molecule, which is highly variable (microheterogeneity). It is obvious that there are differences between the different glycan forms as far as the antiinflammatory property of the protein is concerned. Together with data in the literature, the results presented here suggest a variety of potential indications for therapeutic use of alpha 1-acid glycoprotein in humans.


Sujet(s)
Orosomucoïde/pharmacologie , Animaux , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Encéphale/effets des médicaments et des substances chimiques , Perméabilité capillaire/effets des médicaments et des substances chimiques , Angiopathies intracrâniennes/complications , Évaluation préclinique de médicament , Oedème/étiologie , Oedème/prévention et contrôle , Cochons d'Inde , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Orosomucoïde/usage thérapeutique , Rats
17.
Nephron ; 81(2): 194-9, 1999 Feb.
Article de Anglais | MEDLINE | ID: mdl-9933755

RÉSUMÉ

A minimal change nephrosis was induced in rats by a single intraperitoneal injection of puromycin aminonucleoside (100 mg/kg). This resulted in increased urine protein output, plasma creatinine, blood urine nitrogen, and relative kidney weight. Electronoptically, there was a retraction of the glomerular podocytic foot processes. When human alpha1-acid glycoprotein was injected at 600 mg/kg intravenously on experimental days 6, 7, 8, and 9 into these animals, urine protein output decreased significantly, and the number of podocytic foot processes increased significantly. alpha1-Acid glycoprotein is rich in sialic acid and largely negatively charged. Its therapeutic role in nephrosis, which is characterized by a loss of sialic acid and a loss of negative charge, thereby leading to a loss of permselectivity, is discussed.


Sujet(s)
Néphrose/traitement médicamenteux , Orosomucoïde/pharmacologie , Animaux , Antibiotiques antinéoplasiques , Créatinine/pharmacocinétique , Débit de filtration glomérulaire , Humains , Glomérule rénal/anatomopathologie , Glomérule rénal/ultrastructure , Mâle , Microscopie électronique , Néphrose/induit chimiquement , Puromycine aminonucléoside , Rats , Rat Wistar , Urine
18.
Fundam Clin Pharmacol ; 12(2): 173-81, 1998.
Article de Anglais | MEDLINE | ID: mdl-9565771

RÉSUMÉ

It was the aim of the present study to investigate the effects of the acute phase protein alpha 1-acid glycoprotein in different models of shock. The human plasma preparation used was without effect on mortality in lipopolysaccharide-injected mice when administered in two different doses (1 or 0.33 g/kg i.v.) and according to different treatment schedules. The same preparation significantly increased survival rate (48 h) in rats with septic peritonitis. This effect was seen when alpha 1-acid glycoprotein (200 mg/kg i.v.) was given 15 min prior to and 24 h after cecal puncture. All other dose regimes tested were without significant effect on survival rate. A hemorrhagic/hypovolemic shock model (including a defined trauma) in rats resuscitated with 200 mg/kg alpha 1-acid glycoprotein resulted in significantly higher values of mean arterial blood pressure, cardiac output and stroke volume when compared to corresponding values obtained after resuscitation with Ringer's solution or 200 mg/kg albumin i.v. (free of alpha 1-acid glycoprotein; placebo formulation). Taking all other possible mechanisms of alpha 1-acid glycoprotein into consideration, the partially protective effects of the preparation are explained by enhancing the capillary barrier function and thereby maintaining perfusion of vital organs.


Sujet(s)
Lipopolysaccharides , Orosomucoïde/usage thérapeutique , Péritonite/traitement médicamenteux , Salmonella typhimurium/métabolisme , Sepsie/traitement médicamenteux , Choc hémorragique/traitement médicamenteux , Animaux , Modèles animaux de maladie humaine , Femelle , Hémodynamique/effets des médicaments et des substances chimiques , Humains , Mâle , Souris , Souris de lignée C57BL , Orosomucoïde/pharmacologie , Péritonite/microbiologie , Rats , Rat Sprague-Dawley , Sepsie/microbiologie , Spécificité d'espèce , Analyse de survie
19.
Arzneimittelforschung ; 47(11): 1195-9, 1997 Nov.
Article de Anglais | MEDLINE | ID: mdl-9428972

RÉSUMÉ

Human lys-plasminogen and the corresponding formulation buffer were tested in a rat model of global cerebral ischemia (clamping of both carotid arteries, withdrawal of 5 ml blood for 30 min). The two main parameters, tested in different experimental set-ups, were 1. brain edema (water content) 23.5 h after reperfusion and 2. assessment of neurological deficits 24, 48 and 72 h after reperfusion. In some groups of animals of the first set-up, brains were examined histologically for microvascular fibrin deposits. In a separate group of animals the fibrinolytic plasma activity of rats treated with 500 CU/kg lys-plasminogen was studied. Concerning brain water content lys-plasminogen completely antagonized the formation of brain edema when given with 500 caseolytic Units (CU)/kg i.v. with blood reperfusion and was still effective when given 30 min later. 200 CU/kg i.v. given with blood reperfusion as well as 500 CU/kg i.v. given 60 min after blood reperfusion proved ineffective. In none of the brains investigated microvascular fibrin deposits were found. In experiments with assessment of neurological deficits, animals treated with 500 CU/kg lys-plasminogen i.v. showed almost no disabilities (like sham operated animals) when compared to ischemic (positive) controls which were rather severely handicapped. The formulation buffer of lys-plasminogen, tested in an equivalent volume, was without any effect in both set-ups. No fibrinolytic activity was found in plasma samples of rats up to 240 min after treatment with 500 CU/kg lys-plasminogen i.v. It is concluded from these experiments that human lys-plasminogen has a protective effect in rats against the sequelae of global cerebral ischemia which is not related to the well-known fibrinolytic potential but might be a separate quality.


Sujet(s)
Encéphalopathie ischémique/traitement médicamenteux , Fragments peptidiques/usage thérapeutique , Plasminogène/usage thérapeutique , Animaux , Eau corporelle/métabolisme , Chimie du cerveau/effets des médicaments et des substances chimiques , Oedème cérébral/anatomopathologie , Encéphalopathie ischémique/anatomopathologie , Encéphalopathie ischémique/physiopathologie , Artères carotides/physiologie , Fibrine/métabolisme , Fibrinolytiques/sang , Humains , Mâle , Rats , Rat Sprague-Dawley , Lésion d'ischémie-reperfusion/anatomopathologie
20.
Arch Int Pharmacodyn Ther ; 331(3): 313-21, 1996.
Article de Anglais | MEDLINE | ID: mdl-9125002

RÉSUMÉ

Anesthetized guinea-pigs were intravenously injected with Evans blue. After intracutaneous injection of agonists (lys-plasminogen, histamine, platelet-activating factor, thrombin, bradykinin), the resulting wheals appeared blue in a dose-dependent manner, due to an enhanced capillary permeability, alpha 1-Acid glycoprotein, given i.v. in different doses (3.125-50 mg/kg) and at different times (30-180 min) before Evans blue administration, antagonized the effects of all agonists listed above. This was shown by a parallel shift of the agonist dose-response curves to the right. The effect was time-dependent (tmax: mainly 120 min) and dose-dependent. alpha 1-Acid glycoprotein antagonized the agonists in the following order: lys-plasminogen > histamine = platelet-activating factor > thrombin > bradykinin. As all agonist mentioned are suggested to play a major role in the shock-related increase in vascular permeability, a putatively beneficial role of alpha1-acid glycoprotein in shock is discussed.


Sujet(s)
Perméabilité capillaire/effets des médicaments et des substances chimiques , Orosomucoïde/pharmacologie , Peau/vascularisation , Animaux , Relation dose-effet des médicaments , Antagonisme des médicaments , Bleu d'Evans , Cochons d'Inde , Histamine/pharmacologie , Mâle , Plasminogène/pharmacologie , Peau/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...