Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 422: 115557, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33932462

ABSTRACT

CSL112 (apolipoprotein A-I, apo AI [human]) is an investigational drug in Phase 3 development for risk reduction of early recurrent cardiovascular events following an acute myocardial infarction (AMI). Although CSL112 is known to be well tolerated with a regimen of four weekly 6 g intravenous infusions after AMI, high doses of reconstituted apo AI preparations can transiently elevate liver enzymes in rats, raising the possibility of additive liver toxicity and toxicokinetic (TK) effects upon co-administration with cholesterol-lowering drugs, i.e., HMG-CoA reductase and proprotein convertase subtilisin/kexin type 9 inhibitors. We performed a toxicity and TK study in CD rats assigned to eleven treatment groups, including two dose levels of intravenous (IV) CSL112 (140 mg/kg, low-dose; 600 mg/kg, high-dose) administered as a single dose, alone or with intravenous alirocumab 50 mg/kg/week and/or oral atorvastatin 10 mg/kg/day. In addition, control groups of atorvastatin and alirocumab alone and in combination were investigated. Results showed some liver enzyme elevations (remaining <2-fold of baseline) related to administration of CSL112 alone. There was limited evidence of an additive effect of CSL112 on liver enzymes when combined, at either dose level, with alirocumab and/or atorvastatin, and histology revealed no evidence of an increased incidence or severity of hepatocyte vacuolation compared to the control treatments. Co-administration of the study drugs had minimal effect on their respective exposure levels, and on levels of total cholesterol and high-density lipoprotein cholesterol. These data support concomitant use of CSL112 with alirocumab and/or atorvastatin with no anticipated negative impact on liver safety and TK.


Subject(s)
Antibodies, Monoclonal, Humanized/toxicity , Anticholesteremic Agents/toxicity , Atorvastatin/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Lipoproteins, HDL/toxicity , Liver/drug effects , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Anticholesteremic Agents/pharmacokinetics , Atorvastatin/pharmacokinetics , Biomarkers/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cholesterol/blood , Drug Interactions , Female , Lipoproteins, HDL/pharmacokinetics , Liver/metabolism , Liver/pathology , Male , Rats, Sprague-Dawley , Risk Assessment , Toxicity Tests , Toxicokinetics
2.
Eur J Pharmacol ; 820: 206-216, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29274331

ABSTRACT

New therapeutic agents are needed to overcome the toxicity and suboptimal efficacy observed in current treatment of glomerulonephritis (GN). BaxB01 is a fully human monoclonal antibody targeting a disease-related immunologically distinct isoform of Macrophage migration Inhibitory Factor (MIF), designated oxidized MIF (oxMIF) and locally expressed in inflammatory conditions. We report the pharmacokinetic profile of BaxB01, and its dose and exposure-related disease-modifying activity in experimentally induced rat GN. BaxB01 bound to rat oxMIF with high affinity and reduced rat macrophage migration in vitro. After intravenous administration in rats, BaxB01 demonstrated favorable pharmacokinetics, with a half-life of up to nine days. Disease modification was dose-related (≥ 10mg/kg) as demonstrated by significantly reduced proteinuria and diminished histopathological glomerular crescent formation. Importantly, a single dose was sufficient to establish an exposure-related, anti-inflammatory milieu via amelioration of glomerular cellular inflammation. Pharmacodynamic modeling corroborated these findings, consistently predicting plasma exposures that were effective in attenuating both anti-inflammatory activity and reducing loss of kidney function. This pharmacologic benefit on glomerular function and structure was sustained during established disease, while correlation analyses confirmed a link between the antibody's anti-inflammatory activity and reduced crescent formation in individual rats. Finally, safety assessment in rats showed that the experimental therapeutic was well tolerated without signs of systemic toxicity or negative impact on kidney function. These data define therapeutically relevant exposures correlated with mechanism-based activity in GN, while toxicological evaluation suggests a large therapeutic index and provides evidence for achieving safe and effective exposure to a MIF isoform-directed therapeutic in nephritis-associated disease.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Glomerulonephritis/drug therapy , Glomerulonephritis/immunology , Macrophage Migration-Inhibitory Factors/immunology , Molecular Targeted Therapy , Safety , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Progression , Female , Glomerulonephritis/metabolism , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Male , Monocytes/cytology , Monocytes/drug effects , Protein Isoforms/immunology , Rats
3.
Article in English | MEDLINE | ID: mdl-28775711

ABSTRACT

The hypothalamus-pituitary-thyroid feedback control is a dynamic, adaptive system. In situations of illness and deprivation of energy representing type 1 allostasis, the stress response operates to alter both its set point and peripheral transfer parameters. In contrast, type 2 allostatic load, typically effective in psychosocial stress, pregnancy, metabolic syndrome, and adaptation to cold, produces a nearly opposite phenotype of predictive plasticity. The non-thyroidal illness syndrome (NTIS) or thyroid allostasis in critical illness, tumors, uremia, and starvation (TACITUS), commonly observed in hospitalized patients, displays a historically well-studied pattern of allostatic thyroid response. This is characterized by decreased total and free thyroid hormone concentrations and varying levels of thyroid-stimulating hormone (TSH) ranging from decreased (in severe cases) to normal or even elevated (mainly in the recovery phase) TSH concentrations. An acute versus chronic stage (wasting syndrome) of TACITUS can be discerned. The two types differ in molecular mechanisms and prognosis. The acute adaptation of thyroid hormone metabolism to critical illness may prove beneficial to the organism, whereas the far more complex molecular alterations associated with chronic illness frequently lead to allostatic overload. The latter is associated with poor outcome, independently of the underlying disease. Adaptive responses of thyroid homeostasis extend to alterations in thyroid hormone concentrations during fetal life, periods of weight gain or loss, thermoregulation, physical exercise, and psychiatric diseases. The various forms of thyroid allostasis pose serious problems in differential diagnosis of thyroid disease. This review article provides an overview of physiological mechanisms as well as major diagnostic and therapeutic implications of thyroid allostasis under a variety of developmental and straining conditions.

4.
Expert Rev Clin Pharmacol ; 8(2): 163-77, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25660348

ABSTRACT

Nonacog gamma is a new recombinant factor IX to treat factor IX deficiency. It is indicated for control of bleeding episodes, perioperative management and routine prophylaxis to prevent or reduce the frequency of bleeding episodes in adults and children with hemophilia B. Nonacog gamma was first approved in the USA in June 2013 under the trade name RIXUBIS followed by market approvals in Australia and the EU in 2014, and marketing authorization decision is pending in Japan. Nonacog gamma is derived from a recombinant Chinese hamster ovary cell line using a state of the art biotechnological manufacturing process. Recombinant factor IX is produced by Baxter's protein-free fermentation technology, which was first developed for ADVATE. The product is purified and formulated in the absence of any human or animal-derived protein. Nonacog gamma was characterized both in comprehensive in vitro and in vivo non-clinical studies as well as in an extensive clinical trial program.


Subject(s)
Factor IX/therapeutic use , Hemophilia B/drug therapy , Hemostatics/therapeutic use , Adult , Animals , CHO Cells , Child , Cricetinae , Cricetulus , Humans , Recombinant Proteins/therapeutic use
5.
Int J Hematol ; 98(5): 525-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24061775

ABSTRACT

Baxter has developed a new recombinant factor IX (rFIX) drug product (BAX326) for treating patients with hemophilia B, or congenital FIX deficiency. An extensive preclinical program evaluated the pharmacokinetics, efficacy, and safety of BAX326 in different species. The efficacy of BAX326 was tested in three mouse models of primary pharmacodynamics: tail-tip bleeding, carotid occlusion, and thrombelastography. The pharmacokinetics was evaluated after a single intravenous bolus injection in mice, rats, and macaques. Toxicity was assessed in rats and macaques, safety pharmacology in rabbits and macaques, and immunogenicity in mice. BAX326 was shown to be efficacious in all three primary pharmacodynamic studies (P ≤ 0.0076). Hemostatic efficacy was dose related and similar for the three lots tested. Pharmacokinetic results showed that rFIX activity and rFIX antigen concentrations declined in a bi-phasic manner, similar to a previously licensed rFIX product. BAX326 was well tolerated in rabbits and macaques at all dose levels; no thrombogenic events and no adverse clinical, respiratory, or cardiovascular effects occurred. BAX326 was also shown to have a similar immunogenicity profile to the comparator rFIX product in mice. These results demonstrate that BAX326 has a favorable preclinical safety and efficacy profile, predictive of a comparable effect to that of the previously licensed rFIX in humans.


Subject(s)
Factor IX/pharmacology , Hemophilia B/drug therapy , Recombinant Proteins/pharmacology , Animals , Bleeding Time , Blood Coagulation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Factor IX/administration & dosage , Factor IX/adverse effects , Hemophilia B/blood , Humans , Macaca , Male , Mice , Rabbits , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Thrombelastography , Treatment Outcome
6.
Blood ; 120(26): 5217-23, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-22915644

ABSTRACT

Coronary heart disease is a major cause of death in the western world. Although essential for successful recovery, reperfusion of ischemic myocardium is inevitably associated with reperfusion injury. To investigate a potential protective role of ADAMTS13, a protease cleaving von Willebrand factor multimers, during myocardial ischemia/reperfusion, we used a mouse model of acute myocardial infarction. We found that Adamts13(-/-) mice developed larger myocardial infarctions than wild-type control mice, whereas treatment of wild-type mice with recombinant human ADAMTS13 (rhADAMTS13) led to smaller infarctions. The protective effect of ADAMTS13 was further confirmed by a significant reduction of cardiac troponin-I release and less myocardial apoptosis in mice that received rhADAMTS13 compared with controls. Platelets adherent to the blood vessel wall were observed in few areas in the heart samples from mice treated with vehicle and were not detected in samples from mice treated with rhADAMTS13. However, we observed a 9-fold reduction in number of neutrophils infiltrating ischemic myocardium in mice that were treated with rhADAMTS13, suggesting a potent anti-inflammatory effect of ADAMTS13 during heart injury. Our data show that ADAMTS13 reduces myocardial ischemia/reperfusion injury in mice and indicate that rhADAMTS13 could be of therapeutic value to limit myocardial ischemia/reperfusion injury.


Subject(s)
ADAM Proteins/pharmacology , Anti-Inflammatory Agents/pharmacology , Cardiotonic Agents/pharmacology , Cytoprotection/drug effects , Myocardial Reperfusion Injury/prevention & control , ADAM Proteins/adverse effects , ADAM Proteins/pharmacokinetics , ADAM Proteins/therapeutic use , ADAMTS13 Protein , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , CHO Cells , Cardiotonic Agents/adverse effects , Cardiotonic Agents/pharmacokinetics , Cricetinae , Cricetulus , Cytoprotection/genetics , Humans , Male , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Metalloendopeptidases/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
7.
Blood ; 119(25): 6128-35, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22529289

ABSTRACT

Deficiency of a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), a VWF-cleaving protease, is the key factor in the pathogenesis of thrombotic thrombocytopenic purpura (TTP), a life-threatening thrombotic microangiopathy. It is well established that ADAMTS13 deficiency results in elevated plasma levels of ultra-large VWF multimers (ULVWF), which are prone to induce platelet aggregation; however, the actual trigger of TTP development remains uncertain. Here we describe a new animal model in which some TTP-like symptoms can be triggered in ADAMTS13 knockout mice by challenge with 2000 units/kg body weight of recombinant human VWF containing ULVWF multimers. Animals rapidly showed clinical symptoms and developed severe thrombocytopenia. Schistocytosis, a decrease in hematocrit, and elevated serum lactate dehydrogenase levels were observed. The heart was identified as the most sensitive target organ with rapid onset of extensive platelet aggregation in the ventricles and myocardial necrosis. Prophylactic administration of 200 units/kg recombinant human ADAMTS13 protected ADAMTS13 knockout mice from developing TTP. Therapeutic administration of 320 units/kg rhADAMTS13 reduced the incidence and severity of TTP findings in a treatment interval-dependent manner. We therefore consider this newly established mouse model of thrombotic microangiopathy highly predictive for investigating the efficacy of new treatments for TTP.


Subject(s)
ADAM Proteins/therapeutic use , Disease Models, Animal , Metalloendopeptidases/genetics , Mice, Knockout , Purpura, Thrombotic Thrombocytopenic/drug therapy , Purpura, Thrombotic Thrombocytopenic/genetics , ADAM Proteins/administration & dosage , ADAMTS13 Protein , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Female , Hematocrit , Humans , Male , Mice , Mice, Inbred C57BL , Platelet Count , Purpura, Thrombotic Thrombocytopenic/blood , Purpura, Thrombotic Thrombocytopenic/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Treatment Outcome
8.
Semin Thromb Hemost ; 36(5): 522-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20632249

ABSTRACT

Von Willebrand factor (VWF) is cleaved by the plasma metalloprotease ADAMTS13 ( A Disintegrin and Metalloproteinase with Thrombo Spondin repeats, number 13) that regulates the hemostatic activity of VWF by limiting its multimeric size in the human system. In vitro and ex vivo studies have shown that human recombinant VWF (rVWF) is virtually resistant to the proteolytic activity of murine ADAMTS13. In contrast, rabbit and cynomolgus ADAMTS13 is able to cleave human rVWF. These findings were consistent with in vivo results showing distinct pharmacological behavior of human rVWF depending on the cleaving capacity of ADAMTS13 present in the species tested. Studies were performed using three mouse strains (ADAMTS13 deficient, C57BL/6J [wild type], VWF deficient), rats, rabbits, and cynomolgus monkeys. All animals were infused once with different doses of human rVWF and, in addition, 14 daily doses were given to rats and cynomolgus monkeys. Exaggerated pharmacological effects were observed in mice, with the ADAMTS13 knockout mouse being the most sensitive strain. Similar findings with decreased incidence and severity were seen in normal C57BL/6J mice and also in VWF-deficient mice, where they were least pronounced. In rats, exaggerated pharmacological effects were observed only after 14 doses. Rabbits and cynomolgus monkeys showed no exaggerated pharmacological effects. These differences between species and between mouse strains suggest that the efficiency of ADAMTS13 to cleave rVWF determines the severity of clinical, laboratory and pathohistological findings. These observations highlight the importance of evaluating species' suitability for the generation of meaningful preclinical data for determining the therapeutic safety margins for human patients. Only animals with a sufficient rVWF cleavage capacity by endogenous ADAMTS13 (rabbits and cynomolgus monkeys) are considered appropriate animal models for preclinical evaluation of the rVWF product.


Subject(s)
ADAM Proteins/metabolism , von Willebrand Factor/pharmacology , Animals , Humans , Recombinant Proteins/pharmacology
9.
Toxicol Appl Pharmacol ; 240(1): 117-22, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19540255

ABSTRACT

Statistical comparison of organ weights between treated and untreated animals have traditionally been used to predict potential toxicity for patients. The manner of presentation of organ weight data, and the value of statistical analyses have been topics of discussion. Historically, a decision tree approach has been applied for statistical comparison of organ weights which does not control the overall error rate and can lead to different statistical tests being used by chance for identical settings causing confusion. This paper proposes a simple nonparametric approach for assessing treatment effects on organ weights in terms of ratios based on the Hodges-Lehmann estimator. This allows for simple interpretation of results and aids in the identification of potential target organs as the evaluation is based on effect sizes and not on p-values allowing a robust proof of effect as well as a robust proof of no effect. The proposed estimate and the corresponding nonparametric confidence interval applied to a rank-sum score can be used as a confirmatory test for difference and as a confirmatory test for equivalence. Exploratory analyses can be performed calculating the proposed estimates for each organ separately to be summarized graphically in a confidence interval plot.


Subject(s)
Statistics as Topic , Toxicology/methods , Toxicology/statistics & numerical data , Animals , Confidence Intervals , Humans , Organ Size/drug effects , Organ Size/physiology , Reproducibility of Results , Statistics, Nonparametric
10.
J Virol ; 83(10): 5192-203, 2009 May.
Article in English | MEDLINE | ID: mdl-19279103

ABSTRACT

The timely development of safe and effective vaccines against avian influenza virus of the H5N1 subtype will be of the utmost importance in the event of a pandemic. Our aim was first to develop a safe live vaccine which induces both humoral and cell-mediated immune responses against human H5N1 influenza viruses and second, since the supply of embryonated eggs for traditional influenza vaccine production may be endangered in a pandemic, an egg-independent production procedure based on a permanent cell line. In the present article, the generation of a complementing Vero cell line suitable for the production of safe poxviral vaccines is described. This cell line was used to produce a replication-deficient vaccinia virus vector H5N1 live vaccine, dVV-HA5, expressing the hemagglutinin of a virulent clade 1 H5N1 strain. This experimental vaccine was compared with a formalin-inactivated whole-virus vaccine based on the same clade and with different replicating poxvirus-vectored vaccines. Mice were immunized to assess protective immunity after high-dose challenge with the highly virulent A/Vietnam/1203/2004(H5N1) strain. A single dose of the defective live vaccine induced complete protection from lethal homologous virus challenge and also full cross-protection against clade 0 and 2 challenge viruses. Neutralizing antibody levels were comparable to those induced by the inactivated vaccine. Unlike the whole-virus vaccine, the dVV-HA5 vaccine induced substantial amounts of gamma interferon-secreting CD8 T cells. Thus, the nonreplicating recombinant vaccinia virus vectors are promising vaccine candidates that induce a broad immune response and can be produced in an egg-independent and adjuvant-independent manner in a proven vector system.


Subject(s)
Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Chlorocebus aethiops , Defective Viruses/genetics , Female , Influenza A Virus, H5N1 Subtype/genetics , Interferon-gamma/analysis , Mice , Mice, Inbred BALB C , Mice, Nude , Orthomyxoviridae Infections/immunology , Vaccinia virus/genetics , Vero Cells , Virus Cultivation
11.
Br J Pharmacol ; 139(5): 989-97, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12839873

ABSTRACT

1. Glycerol trinitrate (GTN) has been used in therapy for more than 100 years. Biological effects of GTN are due to the release of the biomediator nitric oxide (NO). However, the mechanism by which GTN provides NO, in particular in liver, is still unknown. In this study, we provide experimental evidence showing that cytoplasm, endoplasmic reticulum, and mitochondria are required for the release of NO from GTN in the liver. 2. NO and nitrite (NO(2)(-)) were determined using low-temperature electron paramagnetic resonance and the Griess reaction, respectively. 3. The first step of GTN biotransformation is the release of NO(2)(-). This step is performed in cytoplasm and catalyzed by glutathione-S-transferase. The second step is the rate-limiting step where NO(2)(-) is slowly reduced to NO. This is mainly catalyzed by cytochrome P-450. The second phase can be significantly enhanced by decreasing the pH value, a situation which occurs during ischemia. At high NADPH concentrations exceeding physiological values, cytochrome P-450 catalyzes GTN biotransformation without the involvement of cytoplasmic glutathione-S-transferase. 4. In conclusion, our data show that NO(2)(-) derived from the first step of biotransformation of GTN in the liver is the precursor of NO but not a product of NO degradation; consequently, NO(2)(-) levels are not likely to be a marker of NO release from GTN as earlier suggested.


Subject(s)
Intracellular Fluid/metabolism , Liver/metabolism , Nitric Oxide/metabolism , Nitroglycerin/metabolism , Animals , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , Male , Mitochondria/metabolism , Nitrites/metabolism , Rats , Rats, Sprague-Dawley
12.
Biofactors ; 19(1-2): 79-85, 2003.
Article in English | MEDLINE | ID: mdl-14757980

ABSTRACT

Here we show that lipid peroxidation of liposomal membranes was suppressed in the presence of Caroverine, a spasmolytic drug used in some countries. In order to understand the mechanism of this antioxidant action of Caroverine we studied the interaction of Caroverine with superoxide radicals, hydroxyl radicals and peroxynitrite. The results of the study show that the reaction of Caroverine with O2-* radicals is of marginal significance. However, it is efficient in removing peroxynitrite and a particular high reaction constant was found for reaction with hydroxyl radicals. The strong antioxidant activity of Caroverine is therefore based both on the partial prevention of the formation and the highly active scavenging of hydroxyl radicals.


Subject(s)
Antioxidants/chemistry , Lipid Peroxidation/drug effects , Quinoxalines/chemistry , Analysis of Variance , Antioxidants/pharmacology , Hydroxyl Radical/chemistry , Hydroxyl Radical/metabolism , Iron/chemistry , Liposomes/chemistry , Liposomes/metabolism , Peroxynitrous Acid/chemistry , Peroxynitrous Acid/metabolism , Quinoxalines/pharmacology , Glycine max/chemistry , Glycine max/metabolism , Superoxides/chemistry , Superoxides/metabolism , Time Factors , Vitamin E/chemistry , Vitamin E/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...