Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Xenobiotica ; 52(5): 453-462, 2022 May.
Article in English | MEDLINE | ID: mdl-35616579

ABSTRACT

Vericiguat is a soluble guanylate cyclase stimulator. The pharmacokinetics, absorption, metabolism, and excretion properties of vericiguat in rats and dogs and the distribution in rats are reported. [14C]-labelled vericiguat was studied in intact and bile duct-cannulated rats (oral and intravenous administration), and dogs (oral administration).Vericiguat reached maximum plasma concentrations at 1-3 h after oral administration. Absolute bioavailability was moderate in rats and high in dogs. Vericiguat was the most abundant component in plasma of rats and dogs.After oral administration to rats, radioactivity was widely distributed. Penetration into the brain was minimal. Elimination was rapid from most tissues in rats. Most of the radioactivity was excreted in faeces (rat: 81%, dog: 89%), while low amounts were excreted in urine (rat: 11%, dog: 4%). Clearance routes in both species were unchanged excretion and metabolism via glucuronidation and oxidative reactions. After intravenous administration to bile duct-cannulated rats, a relevant proportion of the dose (30%) underwent direct excretion into the gastrointestinal tract as unchanged vericiguat.


Subject(s)
Heterocyclic Compounds, 2-Ring , Pyrimidines , Administration, Oral , Animals , Dogs , Feces , Injections, Intravenous , Rats , Tissue Distribution
2.
Xenobiotica ; 50(8): 967-979, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32003293

ABSTRACT

1. Darolutamide is a novel selective androgen receptor antagonist consisting of two pharmacologically equipotent diastereoisomers. The absorption, distribution, metabolism and excretion properties of darolutamide in rats are reported.2. Non- or [14C]-labelled darolutamide, its diastereoisomers and major metabolite were studied in intact and bile duct-cannulated rats (oral and intravenous administration), and rat hepatocytes.3. Darolutamide was quickly (1 h to reach maximum plasma concentration) and completely absorbed after oral administration. Absolute bioavailability was high. Keto-darolutamide was the most abundant metabolite in rat hepatocytes and the only major one in plasma. Interconversion between diastereoisomers was observed.4. After oral administration, radioactivity distributed widely and homogeneously. Penetration into brain was low (brain/blood ratio = 0.079). Elimination was rapid from most tissues. Excretion occurred rapidly, and routes were similar irrespective of administration routes. Complete mass balance was reached by 168 h post-dose. Most radioactivity (61-64%) was excreted in faeces, while relevant amounts (30-33%) were also excreted into urine. The main clearance routes were metabolism via oxidative reactions and glucuronidation. After intravenous administration, a relevant extent of the dose (20%) underwent extrabiliary excretion as darolutamide.


Subject(s)
Androgen Receptor Antagonists/pharmacokinetics , Pyrazoles/pharmacokinetics , Administration, Oral , Animals , Bile/metabolism , Biological Availability , Body Fluids , Feces , Intestinal Absorption , Rats , Tissue Distribution
3.
Eur J Pharm Sci ; 130: 11-20, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30654111

ABSTRACT

The covalent binding of proteins with polyethylene glycol (PEG) molecules is a valuable tool to extend the half-life of many biotherapeutics, including factor VIII (FVIII) products to treat patients with haemophilia A. Although PEG has low toxicity, accumulation of large PEG molecules (>20-30 kDa) with long-term exposure is a potential concern. Thus, it is important to determine whether sufficient excretion processes exist for PEG molecules used in biotherapeutics. BAY 94-9027 is an extended-half-life FVIII product modified through addition of a 60-kDa (branched: dual 30-kDa) PEG molecule. BAY 1025662 is the 60-kDa PEG moiety used for PEGylation of BAY 94-9027. This study investigated the pharmacokinetic (PK) properties, distribution, and excretion of BAY 1025662 in rats in order to predict estimated 60-kDa PEG PK properties in patients. Plasma concentrations in male rats after a single 11-mg/kg intravenous dose of BAY 1025662 (approximating the cumulative PEG-60 exposure in patients during 30 years of BAY 94-9027 treatment) decreased with an initial half-life of 119 h (5 days) in the interval of 114-336 h post administration. Single-dose mass balance studies using radiolabeled BAY 1025662 ([prop-14C]BAY 1025662) showed that 30.4% of radioactivity was excreted within 1 week and 79.1% by Day 168 (primarily in urine). The terminal half-life of radioactivity elimination was approximately 24 days in blood and plasma and was 31-68 days in the majority of other organs up to Day 168. Elimination was nearly complete at the end of the experiment on Day 168; only ~4% of residual radioactivity was present in the animal body. There was no irreversible binding of radioactivity to any tissues and no penetration of the blood-brain barrier. Based on these results, very low steady-state concentrations of 60-kDa PEG were predicted in patients treated with BAY 94-9027, and the validity of these predictions was supported by clinical studies in which almost all 179 patients receiving BAY 94-9027 for prophylaxis had undetectable PEG in plasma for up to >5 years; those with detectable PEG levels demonstrated concentrations within the predicted range. These combined preclinical and clinical observations suggest that excretion processes are in place for high-molecular-weight PEGs such as the PEG-60 moiety used in BAY 94-9027.


Subject(s)
Factor VIII/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Recombinant Proteins/pharmacokinetics , Animals , Autoradiography/methods , Factor VIII/metabolism , Factor VIII/urine , Forecasting , Humans , Male , Polyethylene Glycols/metabolism , Rats , Rats, Wistar , Recombinant Proteins/blood , Recombinant Proteins/urine , Species Specificity , Tissue Distribution/drug effects , Tissue Distribution/physiology
4.
J Pharmacol Exp Ther ; 338(1): 372-80, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21515813

ABSTRACT

Rivaroxaban, an oral, direct factor Xa inhibitor, has a dual mode of elimination in humans, with two-thirds metabolized by the liver and one-third renally excreted unchanged. P-glycoprotein (P-gp) is known to be involved in the absorption, distribution, and excretion of drugs. To investigate whether rivaroxaban is a substrate of P-gp, the bidirectional flux of rivaroxaban across Caco-2, wild-type, and P-gp-overexpressing LLC-PK1 cells was investigated. Furthermore, the inhibitory effect of rivaroxaban toward P-gp was determined. Rivaroxaban exhibited high permeability and polarized transport across Caco-2 cells. Rivaroxaban was shown to be a substrate for, but not an inhibitor of, P-gp. Of a set of potential P-gp inhibitors, ketoconazole and ritonavir, but not clarithromycin or erythromycin, inhibited P-gp-mediated transport of rivaroxaban, with half-maximal inhibitory concentration values in the range of therapeutic plasma concentrations. These findings are in line with observed area under the plasma concentration-time curve increases in clinical drug-drug interaction studies indicating a possible involvement of P-gp in the distribution and excretion of rivaroxaban. In vivo studies in wild-type and P-gp double-knockout mice demonstrated that the impact of P-gp alone on the pharmacokinetics of rivaroxaban is minor. However, in P-gp double-knockout mice, a slight increase in brain concentrations and decreased excretion into the gastrointestinal tract were observed compared with wild-type mice. These studies also demonstrated that brain penetration of rivaroxaban is fairly low. In addition to P-gp, a further transport protein might be involved in the secretion of rivaroxaban.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Morpholines/metabolism , Thiophenes/metabolism , Animals , Caco-2 Cells , Humans , LLC-PK1 Cells , Male , Mice , Mice, Knockout , Morpholines/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Rivaroxaban , Swine , Thiophenes/pharmacology
5.
Blood Press ; 15(2): 116-28, 2006.
Article in English | MEDLINE | ID: mdl-16754275

ABSTRACT

This study compares the effect of two different strategies to inhibit the renin-angiotensin system in the setting of acute myocardial infarction (MI). Male Wistar rats were treated with placebo, the angiotensin-converting enzyme (ACE) inhibitor ramipril (1 mg/kg/day), or the AT1 receptor antagonist, olmesartan (1 mg/kg/day), both initiated 1 week before induction of MI and continued for 6 weeks after MI. The inflammatory reaction in the heart was investigated 7 days post-MI by determination of macrophage infiltration and the expression of tumor necrosis factor (TNF-alpha), interleukin (IL)-1beta and IL-6 at mRNA and protein levels. Six weeks post-MI, cardiac function was measured following chronic implantation of catheters in the LV and femoral artery, and cardiac morphology and coronary structure were investigated in picrosirius-red stained hearts. In placebo-treated rats, macrophage infiltration was accompanied by upregulation of IL-1beta and IL-6 mRNA in the peri-infarct zone. TNF-alpha and IL-1beta mRNA and protein were also upregulated in the non-infarcted myocardium. Whereas both treatment regimes significantly reduced IL-6 upregulation, olmesartan additionally reduced macrophage infiltration and IL-1beta expression. Six weeks post-MI, placebo-treated MI animals developed an impaired cardiac function with structural remodeling of the myocardium and coronaries. While olmesartan and ramipril both improved cardiac function and reduced infarct size and myocardial/coronary remodeling, olmesartan was more effective not only in increasing vascular perimeter, inner vascular diameter and septal thickness but also in lowering media thickness of coronary arteries, inner left ventricular diameter, left ventricular circumference and left ventricular end-diastolic pressure than ramipril. Thus, following MI the AT1 receptor blocker, olmesartan, attenuated cardiac inflammatory reactions and protected myocardial/coronary structure and function of the failing heart proving to be of similar, in some cases superior effectiveness in this respect than the ACE inhibitor, ramipril.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Imidazoles/therapeutic use , Inflammation/drug therapy , Myocardial Infarction/drug therapy , Ramipril/therapeutic use , Tetrazoles/therapeutic use , Acute Disease , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular System/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Inflammation/immunology , Interleukin-1/biosynthesis , Interleukin-1/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Male , Myocardial Infarction/immunology , Placebos , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , Rats , Rats, Wistar , Renin-Angiotensin System/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
6.
CNS Drug Rev ; 9(4): 343-58, 2003.
Article in English | MEDLINE | ID: mdl-14647528

ABSTRACT

Traumatic brain injury (TBI) is the most common cause of mortality and morbidity in adults under 40 years of age in industrialized countries. Worldwide the incidence is increasing, about 9.5 million people are hospitalized per year due to TBI, and the death rate is estimated to be more than one million people per year. Recently BAY 38-7271 has been characterized as a structurally novel, selective and highly potent cannabinoid CB1/CB2 receptor agonist in vitro and in vivo with pronounced neuroprotective efficacy in a rat traumatic brain injury model, showing a therapeutic window of at least 5 h. Furthermore, neuroprotective efficacy was also found in models of transient and permanent occlusion of the middle cerebral artery and brain edema models as well. In this article we review the in vitro and in vivo pharmacology of BAY 38-7271, the results from acute and subacute toxicity studies, pharmacokinetics and drug metabolism in animals and healthy male volunteers. In phase I studies BAY 38-7271 was safe and well tolerated when administered by i.v. infusion for either 1 or 24 h. As the doses of BAY 38-7271 in animals needed for maximal neuroprotective efficacy were significantly lower than those inducing typical cannabinoid-like side effects, it is to be expected that the compound will offer a novel therapeutic approach with a favorable therapeutic window for the treatment of TBI or cerebral ischemia.


Subject(s)
Brain Injuries/drug therapy , Cannabinoid Receptor Agonists , Indans/therapeutic use , Neuroprotective Agents/therapeutic use , Sulfonic Acids/therapeutic use , Animals , Area Under Curve , Half-Life , Humans , Indans/metabolism , Indans/pharmacokinetics , Metabolic Clearance Rate , Sulfonic Acids/metabolism , Sulfonic Acids/pharmacokinetics
7.
Eur J Pharmacol ; 458(1-2): 3-16, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12498901

ABSTRACT

Bradykinin B(1) and B(2) receptors are up-regulated in the infarcted myocardium, and both receptors are involved in the regulation of intracellular pH and Ca(2+). The present study investigated the role of bradykinin B(1) and B(2) receptors in the regulation of Na(+)-H(+) exchanger (NHE-1), Na(+)-Ca(2+) exchanger (NCE-1) and Na(+)-HCO(3)(-) symporter (NBC-1) in the infarcted myocardium. NHE-1, NCE-1 and NBC-1 mRNA expression was determined by Northern blot analysis and the protein levels by Western blot analysis. Measurements were performed 1, 7 and 14 days after induction of myocardial infarction. Localization of NHE-1, NCE-1 and NBC-1 within the myocardium was studied using confocal microscopy. Cardiac morphology was measured in picrosiris-red-stained hearts. Rats were treated with placebo, the bradykinin B(2) receptor antagonist icatibant (0.5 mg/kg/day) or the bradykinin B(1) receptor antagonist des-Arg(9)-[Leu(8)]bradykinin (1 mg/kg/day). Treatment was started 1 week prior to surgery and continued until 1, 7 and 14 days post infarction. NHE-1, NCE-1 and NBC-1 mRNA expression and protein levels were increased 1 day and reached maximum values on day 7 post infarction. NHE-1 was localized in the plasma membrane, NCE-1 in the membrane of the sarcoplasmatic reticulum and NBC-1 near the Z-line. Icatibant reduced NHE-1 and inhibited NCE-1 mRNA- and protein up-regulation, while des-Arg(9)-[Leu(8)]bradykinin had no effect on NHE-1 and NCE-1 expression and translation. Transcriptional and translational up-regulation of NBC-1 was unaffected by the bradykinin B(1) and B(2) receptor antagonists. Icatibant, but not des-Arg(9)-[Leu(8)]bradykinin, limited infarct size and reduced left ventricular dilation, septal thickening and interstitial fibrosis post infarction. Bradykinin B(2) receptors are involved in transcriptional and translational regulation of NHE-1 and NCE-1 in the ischemic myocardium. Chronic B(2) receptor blockade might exert an anti-ischemic effect via limitation of NHE-1-mediated acidosis and NCE-1-mediated Ca(2+)-overload.


Subject(s)
Bradykinin/analogs & derivatives , Ion Pumps/metabolism , Myocardium/metabolism , Receptors, Bradykinin/physiology , Animals , Blotting, Northern , Blotting, Western , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , Immunohistochemistry , Ion Pumps/genetics , Male , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardium/pathology , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Bradykinin B1 , Receptor, Bradykinin B2 , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Transcription, Genetic/drug effects
8.
Eur J Pharmacol ; 453(1): 99-109, 2002 Oct 18.
Article in English | MEDLINE | ID: mdl-12393065

ABSTRACT

Abnormal Ca(2+) inward current through cardiac Ca(2+) channels during ischemia has been shown to be an initial signal for activation of myocardial Ca(2+)-dependent enzymes. This study investigated the contribution of cardiac L- and T-type Ca(2+) channels in the calpain-mediated myocardial damage following myocardial infarction. Myocardial infarction was induced by permanent ligation of the left coronary artery. Infarcted rats were orally treated with placebo, amlodipine (L-channel blockade; 4 mg/kg/day) or mibefradil (L-/T-channel blockade; 10 mg/kg/day) beginning 7 days before induction of myocardial infarction. Gene expression, protein levels and enzyme activity of calpains I and II were measured 1, 3, 7 and 14 days postcoronary occlusion in the noninfarcted and infarcted myocardium. Infarct size, left ventricular dilation and interstitial collagen volume fraction were determined in picrosirius red-stained hearts. Myocardial infarction induced an up-regulation of calpain I mRNA, protein and activity in the noninfarcted myocardium (maximum 14 days postinfarction), whereas mRNA, protein and activity of calpain II were maximally increased in the infarcted myocardium 3 days postinfarction. Fourteen days postinfarction, infarct size was 49%, the left ventricle was dilated and interstitial collagen volume fraction was increased. Amlodipine-inhibited mRNA, protein and activity up-regulation of calpain I decreased interstitial collagen volume fraction and infarct size. Mibefradil-attenuated mRNA, protein and activity up-regulation of calpain II at all four time points measured and of calpain I at 7 and 14 days postinfarction reduced infarct size and prevented left ventricular dilation. Infarction-induced cardiac hypertrophy was accompanied by an up-regulation of calpain I, whereas calpain II was up-regulated in the infarcted myocardium. Cardiac L- and T-type Ca(2+) channel blockade differentially reduced postinfarction remodeling associated with selective inhibition of cardiac calpains I and II, respectively.


Subject(s)
Calcium Channel Blockers/pharmacology , Calpain/biosynthesis , Myocardial Infarction/enzymology , Protein Biosynthesis/drug effects , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Animals , Calpain/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Male , Myocardial Infarction/genetics , Myocardium/enzymology , Protein Biosynthesis/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Transcription, Genetic/physiology , Up-Regulation/physiology
9.
Drugs ; 62 Spec No 1: 43-52, 2002.
Article in French | MEDLINE | ID: mdl-12036388

ABSTRACT

The octapeptide angiotensin II (Ang II), the potent effector molecule of the renin-angiotensin-aldosterone system (RAAS), is involved in the control of blood pressure, cardiac and vascular function as well as sodium and water homeostasis. Because Ang II has also been implicated in the pathophysiology of cardiovascular diseases and renal failure, it has been of increasing interest to inhibit the RAAS at the level of its enzymes such as renin and angiotensin-converting enzyme (ACE) and receptors. At least two subtypes of angiotensin receptors have been identified: AT(1) and AT(2). The AT(1 )receptor mediates all of the known actions of Ang II in the cardiovascular system, such as vasoconstriction, increasing cardiac contractility and renal tubular sodium reabsorption, as well as vascular and cardiac hypertrophy. In contrast, less is known regarding the function of the AT(2) receptor. Evidence suggests that the AT(2) receptor inhibits cell proliferation and induces differentiation, apoptosis and regeneration. The AT(2) receptor has been shown to reverse AT(1) receptor-mediated hypertrophy, suggesting that these receptors exert opposing effects in the cardiovascular system. While renin and ACE inhibitors block the RAAS at the enzymatic level, AT receptor antagonists specifically inhibit the RAAS at the receptor site. AT(1 )receptor antagonists induce a dose-dependent blockade of Ang II-induced effects, resulting in a reduction in blood pressure, cardiac and vascular hypertrophy, proteinuria and glomerular sclerosis. It is postulated that AT(1) receptor antagonists may provide end-organ protection by blocking Ang II via the AT(1) receptor, yet leaving the AT(2) receptor unopposed. These substances have been shown to decrease morbidity and mortality of patients with heart failure and renal disease associated with diabetes.


Subject(s)
Coronary Disease/physiopathology , Heart Failure/physiopathology , Receptors, Angiotensin/metabolism , Renal Insufficiency/physiopathology , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Antihypertensive Agents/pharmacology , Coronary Disease/drug therapy , Heart Failure/drug therapy , Humans , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Renal Insufficiency/drug therapy , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology
10.
Br J Pharmacol ; 135(8): 1951-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11959798

ABSTRACT

1. The calpains have been proposed to be activated following cardiac ischaemia and to contribute to myocyte damage after myocardial infarction (MI). In this study, the activity of calpains I and II in the infarcted and non-infarcted rat myocardium and the action of the selective calpain inhibitor, CAL 9961, has been investigated. 2. MI was induced by permanent ligation of the left coronary artery. One, 3, 7 and 14 days post MI, the enzymes calpain I and II were separated from homogenates of the interventricular septum (IS) and left ventricular free wall (LVFW) by chromatography on DEAE-Sepharose. The activity of the calpains was measured in sham-operated and MI animals chronically treated with placebo or CAL 9961 (15 mg kg(-1) d(-1) s.c.) in a synthetic substrate assay. Treatment was started 3 days before MI induction. 3. Calpain I activity reached highest values in IS 14 days post MI, whereas maximum activity of calpain II was measured in LVFW 3 days post MI. In experiments in vitro, CAL 9961 completely inhibited both calpains. In vivo, chronic treatment of MI animals with CAL 9961 partially prevented the increase in calpain I activity in IS and reduced calpain II activity in LVFW to sham levels. 4. Our findings demonstrate that calpains I and II are activated after MI, however, both enzymes differ in their regional and temporal activation within the infarcted myocardium. Chronic inhibition of these enzymes with CAL 9961 might limit the calpain-induced myocardial damage and preserve cardiac structural integrity post MI.


Subject(s)
Calpain/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Glycoproteins/pharmacology , Myocardial Infarction/drug therapy , Myocardial Infarction/enzymology , Myocardium/enzymology , Animals , Calpain/antagonists & inhibitors , Calpain/isolation & purification , Chronic Disease , Cysteine Proteinase Inhibitors/therapeutic use , Enzyme Activation/drug effects , Glycoproteins/therapeutic use , Male , Myocardial Infarction/pathology , Myocardium/cytology , Myocardium/pathology , Rats , Rats, Wistar , Ventricular Function, Left/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...