Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
1.
J Pharmacol Toxicol Methods ; 60(2): 107-16, 2009.
Article in English | MEDLINE | ID: mdl-19470407

ABSTRACT

Testing for possible cardiovascular side effects of new drugs has been an essential part of drug development for years. A more detailed analysis of the electrocardiogram (ECG) to detect effects on ventricular repolarization (effects on the QT interval), as a marker for possible proarrhythmic potential has been added to that evaluation in recent years. State-of-the art evaluation of drug-induced effects on the QT interval have evolved, but due to the complexity of the assessment, the trend in safety pharmacology studies has been to collect large numbers of high quality ECGs to allow for a robust assessment including the influence of heart rate on the QT interval apart from possible drug-induced effects. Since an assessment of the ECG is often included in toxicological studies, one can consider making such an assessment using ECG data from routine toxicological studies. This review summarizes various aspects of both safety pharmacology and toxicology studies with regards to their impact on the quality and quantity of ECG data that one can reasonably derive. We conclude that ECG data from toxicological studies can offer complementary ECG data that can strengthen a risk assessment. However, for the great majority of standard toxicity studies conducted, the ECG data collected do not permit an adequate assessment of drug-induced effects on the QT interval with the sensitivity expected from the ICH S7B guidelines. Furthermore, sponsors should be discouraged from performing any analyses on low quality ECGs to avoid generating misleading data. Substantial improvements in ECG quality and quantity are available, thereby making a QT interval assessment within the context of a standard toxicological study feasible, but these methods may require a larger commitment of resources from the sponsor. From the viewpoint of risk mitigation and limiting the attrition of promising new therapies, a commitment of resources to insure ECG data quality in either toxicology or safety pharmacology studies may be well justified.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/chemically induced , Electrocardiography/drug effects , Long QT Syndrome/chemically induced , Risk , Safety , Animals , Heart Rate/drug effects , Humans , Toxicity Tests
2.
J Pharm Biomed Anal ; 49(4): 1014-20, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19250790

ABSTRACT

The fully humanized Lewis-Y carbohydrate specific monoclonal antibody (mAb) IGN311 is currently tested in a passive immunotherapy approach in a clinical phase I trail and therefore regulatory requirements demand qualified assays for product analysis. To demonstrate the functionality of its Fc-region, the capacity of IGN311 to mediate complement dependent cytotoxicity (CDC) against human breast cancer cells was evaluated. The "classical" radioactive method using chromium-51 and a FACS-based assay were established and qualified according to ICH guidelines. Parameters evaluated were specificity, response function, bias, repeatability (intra-day precision), intermediate precision (operator-time different), and linearity (assay range). In the course of a fully nested design, a four-parameter logistic equation was identified as appropriate calibration model for both methods. For the radioactive assay, the bias ranged from -6.1% to -3.6%. The intermediate precision for future means of duplicate measurements revealed values from 12.5% to 15.9% and the total error (beta-expectation tolerance interval) of the method was found to be <40%. For the FACS-based assay, the bias ranged from -8.3% to 0.6% and the intermediate precision for future means of duplicate measurements revealed values from 4.2% to 8.0%. The total error of the method was found to be <25%. The presented data demonstrate that the FACS-based CDC is more accurate than the radioactive assay. Also, the elimination of radioactivity and the 'real-time' counting of apoptotic cells further justifies the implementation of this method which was subsequently applied for testing the influence of storage at 4 degrees C and 25 degrees C ('stability testing') on the potency of IGN311 drug product. The obtained results demonstrate that the qualified functional assay represents a stability indicating test method.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies/toxicity , Complement System Proteins/physiology , Lewis Blood Group Antigens/immunology , Algorithms , Biological Assay , Cell Line, Tumor , Cell Survival , Coloring Agents , Data Interpretation, Statistical , Female , Flow Cytometry , Humans , Linear Models , Reproducibility of Results , Risk Assessment
3.
Br J Pharmacol ; 154(7): 1465-73, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18536749

ABSTRACT

BACKGROUND AND PURPOSE: Inhibition of cholesteryl ester transfer protein (CETP) with torcetrapib in humans increases plasma high density lipoprotein (HDL) cholesterol levels but is associated with increased blood pressure. In a phase 3 clinical study, evaluating the effects of torcetrapib in atherosclerosis, there was an excess of deaths and adverse cardiovascular events in patients taking torcetrapib. The studies reported herein sought to evaluate off-target effects of torcetrapib. EXPERIMENTAL APPROACH: Cardiovascular effects of the CETP inhibitors torcetrapib and anacetrapib were evaluated in animal models. KEY RESULTS: Torcetrapib evoked an acute increase in blood pressure in all species evaluated whereas no increase was observed with anacetrapib. The pressor effect of torcetrapib was not diminished in the presence of adrenoceptor, angiotensin II or endothelin receptor antagonists. Torcetrapib did not have a contractile effect on vascular smooth muscle suggesting its effects in vivo are via the release of a secondary mediator. Treatment with torcetrapib was associated with an increase in plasma levels of aldosterone and corticosterone and, in vitro, was shown to release aldosterone from adrenocortical cells. Increased adrenal steroid levels were not observed with anacetrapib. Inhibition of adrenal steroid synthesis did not inhibit the pressor response to torcetrapib whereas adrenalectomy prevented the ability of torcetrapib to increase blood pressure in rats. CONCLUSIONS AND IMPLICATIONS: Torcetrapib evoked an acute increase in blood pressure and an acute increase in plasma adrenal steroids. The acute pressor response to torcetrapib was not mediated by adrenal steroids but was dependent on intact adrenal glands.


Subject(s)
Blood Pressure/drug effects , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Oxazolidinones/toxicity , Quinolines/toxicity , Adrenal Cortex/cytology , Adrenal Cortex/drug effects , Aldosterone/blood , Animals , Anticholesteremic Agents/toxicity , Corticosterone/blood , Dogs , Drug Evaluation, Preclinical , Female , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Models, Animal , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley , Species Specificity
4.
Cardiovasc Res ; 58(1): 32-45, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12667944

ABSTRACT

OBJECTIVE: To attempt to determine the relative value of preclinical cardiac electrophysiology data (in vitro and in vivo) for predicting risk of torsade de pointes (TdP) in clinical use. METHODS: Published data on hERG (or I(Kr)) activity, cardiac action potential duration (at 90% repolarisation; APD(90)), and QT prolongation in dogs were compared against QT effects and reports of TdP in humans for 100 drugs. These data were set against the free plasma concentrations attained during clinical use (effective therapeutic plasma concentrations; ETPC(unbound)). The drugs were divided into five categories: (1) Class Ia and III antiarrhythmics; (2) Withdrawn from market due to TdP; (3) Measurable incidence/numerous reports of TdP in humans; (4) Isolated reports of TdP in humans; (5) No reports of TdP in humans. RESULTS: Data from hERG (or I(Kr)) assays in addition to ETPC(unbound) data were available for 52 drugs. For Category 1 drugs, data for hERG/I(Kr) IC(50), APD(90), QTc in animals and QTc in humans were generally close to or superimposed on the ETPC(unbound) values. This relationship was uncoupled in the other categories, with more complex relationships between the data. In Category 1 (except amiodarone), the ratios between hERG/I(Kr) IC(50) and ETPC(unbound) (max) ranged from 0.1- to 31-fold. Similar ranges were obtained for drugs in Category 2 (0.31- to 13-fold) and Category 3 (0.03- to 35-fold). A large spread was found for Category 4 drugs (0.13- to 35700-fold); this category embraced an assortment of mechanisms ranging from drugs which may well be affecting I(Kr) currents in clinical use (e.g. sparfloxacin) to others such as nifedipine (35700-fold) where channel block is not involved. Finally, for the majority of Category 5 drugs there was a >30-fold separation between hERG/I(Kr) activity and ETPC(unbound) values, with the notable exception of verapamil (1.7-fold), which is free from QT prolongation in man; this is probably explained by its multiple interactions with cardiac ion channels. CONCLUSIONS: The dataset confirms the widely-held belief that most drugs associated with TdP in humans are also associated with hERG K(+) channel block at concentrations close to or superimposed upon the free plasma concentrations found in clinical use. A 30-fold margin between C(max) and hERG IC(50) may suffice for drugs currently undergoing clinical evaluation, but for future drug discovery programmes, pharmaceutical companies should consider increasing this margin, particularly for drugs aimed at non-debilitating diseases. However, interactions with multiple cardiac ion channels can either mitigate or exacerbate the prolongation of APD and QT that would ensue from block of I(Kr) currents alone, and delay of repolarisation per se is not necessarily torsadogenic. Clearly, an integrated assessment of in vitro and in vivo data is required in order to predict the torsadogenic risk of a new candidate drug in humans.


Subject(s)
Anti-Arrhythmia Agents/pharmacokinetics , Drug Evaluation, Preclinical/methods , Electrocardiography/drug effects , Long QT Syndrome/chemically induced , Torsades de Pointes/chemically induced , Animals , Databases, Factual , Death, Sudden, Cardiac/etiology , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Potassium Channels/genetics , Potassium Channels, Voltage-Gated/drug effects , Potassium Channels, Voltage-Gated/genetics , Risk , Safety
5.
Methods Find Exp Clin Pharmacol ; 24(6): 361-70, 2002.
Article in English | MEDLINE | ID: mdl-12224443

ABSTRACT

Endothelin-1 (ET-1) has been found to be one of the most potent vasoconstrictive peptides known, and is therefore considered to be an important factor in diseases such as hypertension, heart failure, pulmonary hypertension, renal diseases, etc. Thus, the development of ET-receptor antagonists may offer a new therapeutic strategy in these fields. In this article, we summarize the method for assessing our compound as a selective ETA-receptor antagonist. Binding assays and in vitro function assays (isolated vessels) were examined for the assessment of in vitro potency, selectivity of the ETA receptor against the ETB receptor, specificity for ET receptors, agonistic activities for ET receptors and the blocking manner of the compound on ET receptors. Chinese hamster ovary (CHO) cells expressing human ET receptors and tissue membrane preparations from both human and animals were used for the binding assays. The specificity of the compound against ET receptors was demonstrated using 116 and 9 receptor binding and enzyme assays, respectively. The agonistic activity and potency of the compound at tissue levels were examined using isolated vessels. We also demonstrated the effect of protein binding on the potency of the compound by adding a physiological concentration of serum albumin to the tissue baths. In vivo potency and features of the compound as a selective ETA-antagonist were confirmed using mice, rats and dogs exogenously treated with ET-1 or big ET-1. We also demonstrated the compound's duration of action and pharmacokinetics in animal models and intact animals, respectively. From these experiments, we found a nonpeptide, potent, orally active and long-lasting, highly selective ETA-receptor antagonist.


Subject(s)
Endothelin Receptor Antagonists , Endothelins/metabolism , Ethers/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Animals , Binding, Competitive , Blood Vessels/drug effects , Blood Vessels/physiology , CHO Cells , Cricetinae , Dogs , Endothelins/pharmacology , Ethers/pharmacokinetics , Humans , Hydrocarbons, Fluorinated/pharmacokinetics , Hypertension/physiopathology , In Vitro Techniques , Rats , Receptor, Endothelin A , Receptor, Endothelin B
6.
J Cardiovasc Pharmacol ; 36(5 Suppl 1): S337-41, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11078414

ABSTRACT

The single oral administration of a mixed endothelin-A-and -B- (ETA/ETB) receptor antagonist, J-104132 (L-753,037) decreased the blood pressure in conscious spontaneously hypertensive rats (SHR), stroke-prone SHR (SHRSP) and Dahl salt-sensitive hypertensive rats fed high-salt diet (DS-H) at doses of 3 and 10 mg/kg, with a duration of approximately 24 h. The magnitude of the antihypertensive effects was greater in DS-H than in SHR and SHRSP Preproendothelin-1 mRNA expression in the kidney and aorta was greater (about twofold) in DS-H than that in nonnotensive Dahl salt-resistant rats fed high-salt diet (DR-H), while there was no difference in preproendothelin-1 mRNA expression between SHR and Wistar Kyoto rats (WKY). An AT1-receptor antagonist, MK-954 (Losartan), also attenuated hypertension in SHR and SHRSP at oral doses of 3 and 10 mg/kg, but bad no effect in DS-H. The concomitant treatment with MK-954 and J-104132 showed additive antihypertensive effects in SHR and SHRSP. In DS-H, MK-954 potentiated the antihypertensive effects of J-104132. From these results, we suggest that: (1) the contribution of endothelin (ET) to the maintenance of hypertension is greater in salt-sensitive hypertensive models than in SHR and SHRSP; (2) the antihypertensive efficacy of ETA/ETB blockade is augmented by AT1-receptor blockade; (3) ET blockers alone or in combination with AT1 antagonists could be useful in the treatment of hypertension for patients who do not respond adequately to renin-angiotensin system inhibitors alone.


Subject(s)
Angiotensin Receptor Antagonists , Antihypertensive Agents/pharmacology , Endothelin Receptor Antagonists , Losartan/pharmacology , Pyridines/pharmacology , Animals , Blood Pressure/drug effects , Drug Synergism , Endothelin-1 , Endothelins/genetics , Male , Protein Precursors/genetics , RNA, Messenger/analysis , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptor, Endothelin A , Receptor, Endothelin B
7.
Hypertension ; 34(3): 514-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10489403

ABSTRACT

The purpose of the present experiment was to study the pathophysiological roles of endothelin-1 (ET-1) in salt-sensitive hypertension with the use of Dahl salt-sensitive (DS) and salt-resistant (DR) rats. PreproET-1 mRNA expression was determined by reverse transcription-polymerase chain reaction. In the kidney, expression of preproET-1 mRNA was greater in DS rats on a normal salt diet compared with DR rats of the same age. In DS rats, the level of preproET-1 mRNA expression in kidney had a significant correlation with systolic blood pressure. The expression of preproET-1 mRNA in aorta and kidney was increased by 3-week high salt intake in DS rats but not in DR rats. Expression of preproET-1 mRNA and ET-1 levels in left ventricle was exaggerated by high salt intake in DS rats. However, there was no significant difference in plasma ET-1 levels between DS and DR rats regardless of salt intake. Pressor response curves for ET-1 in DS rats with or without high salt intake were significantly shifted to the left compared with those in DR rats. A single oral dose (3 to 10 mg/kg) of J-104132 (L-753 037), a potent, orally active mixed endothelin A and B (ET(A)/ET(B)) receptor antagonist, reduced blood pressure to normotensive levels in DS rats with high salt intake, and its action was maintained for >/=24 hours. In DS rats with normal salt intake, J-104132 (10 mg/kg) slightly but significantly decreased blood pressure. DR rats did not show obvious depressor responses to J-104132 (10 mg/kg) regardless of salt intake. These results suggest that ET-1 acts as one of the pathophysiological factors in the development and maintenance of salt-sensitive hypertension, and a mixed ET(A)/ET(B) receptor antagonist could be useful in the treatment for salt-sensitive hypertension.


Subject(s)
Endothelin-1/biosynthesis , Hypertension/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Blood Pressure/drug effects , Endothelin Receptor Antagonists , Endothelin-1/blood , Endothelin-1/genetics , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Myocardium/metabolism , Pressoreceptors/drug effects , Pyridines/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Strains , Receptor, Endothelin A , Receptor, Endothelin B , Ventricular Function, Left
8.
J Pharmacol Exp Ther ; 289(3): 1262-70, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10336515

ABSTRACT

J-104132 [(+)-(5S,6R, 7R)-2-butyl-7-[2-((2S)-2-carboxypropyl)-4-methoxyphenyl]-5-(3, 4-methylenedioxyphenyl)cyclopenteno[1,2-b]pyridine-6-carboxylic; also referred to as L-753,037] is a potent, selective inhibitor of ETA and ETB endothelin (ET) receptors (e.g., Ki: cloned human ETA = 0.034 nM; cloned human ETB = 0.104 nM). In both ligand-binding and isolated tissue preparation protocols, the inhibition of ET receptors with J-104132 is reversible and competitive. In vitro, J-104132 is a potent antagonist of ET-1-induced accumulation of [3H]inositol phosphates in Chinese hamster ovary cells stably expressing cloned human ETA receptors (IC50 = 0.059 nM), ET-1-induced contractions in rabbit iliac artery (pA2 = 9.70) and of BQ-3020-induced contractions in pulmonary artery (pA2 = 10.14). J-104132 is selective for ET receptors because it had no effect on contractions elicited by norepinephrine or KCl in the vascular preparations. The in vivo potency of J-104132 was assessed using challenges with exogenous ET-1. In conscious mice, 5 nmol/kg i.v. ET-1 causes death. Pretreatment with J-104132 prevents the lethal response to ET-1 when administered i.v. (ED50 = 0.045 mg/kg) or p.o. in fed animals (ED50 = 0.35 mg/kg). In conscious, normotensive rats, pressor responses to 0.5 nmol/kg i.v. ET-1 are inhibited by J-104132 after i.v. (0.1 mg/kg) or p.o. (1 mg/kg) administration. In anesthetized dogs, ET-1 was administered directly into the renal artery or brachial artery to generate dose-response (blood flow) curves, and the inhibitory potency of J-104132 (i.v. infusion) was quantified. J-104132 produced greater than 10-fold shifts in the ET-1 dose-response curves at 0.03 mg/kg/h (renal) and 0.3 mg/kg/h (brachial). Oral bioavailability of J-104132 in rats was approximately 40%. These studies indicate that J-104132 is a selective, potent, orally active antagonist of both ETA and ETB receptors and is an excellent pharmacological tool to explore the therapeutic use of a mixed ETA/ETB receptor antagonist.


Subject(s)
Blood Pressure/drug effects , Endothelin Receptor Antagonists , Muscle, Smooth, Vascular/physiology , Pyridines/pharmacology , Animals , Binding, Competitive , CHO Cells , Cloning, Molecular , Cricetinae , Dogs , Endothelin-1/metabolism , Female , Hippocampus/physiology , Humans , Iliac Artery/drug effects , Iliac Artery/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred Strains , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Phosphatidylinositols/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Pyridines/toxicity , Rabbits , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptor, Endothelin B , Receptors, Endothelin/genetics , Recombinant Proteins/antagonists & inhibitors , Renal Artery/drug effects , Renal Artery/physiology , Transfection , Uterus/physiology
9.
Rehabilitation (Stuttg) ; 38(4): 249-53, 1999 Nov.
Article in German | MEDLINE | ID: mdl-10627971

ABSTRACT

OBJECTIVES: Assessment of non-operative treatment of orthopaedic diseases is sometimes considered difficult. The purpose of this retrospective longitudinal study on the one hand is to evaluate the formal quality of questionnaire data under routine conditions in an in-patient rehabilitation hospital and on the other hand to study the impact of additional psychologic treatment on the outcome of patients who received conservative treatment for chronic orthopaedic diseases. PATIENTS AND METHODS: During a 7-month period, a total of 900 patients received a standardized questionnaire at the beginning and the end of a 4-week inhouse treatment for chronic orthopaedic diseases. A questionnaire handed out to the patients at arrival includes 33 items of psychophysiologic interest. At discharge, a modified questionnaire was used, focussing on the course of complaints during the treatment period, and including additional control-questions for consistency. Return of the questionnaires was voluntary, no pressure was applied, no effort was made to correct inconsistent or missing data. The results presented here are based on 105 orthopaedic patients who returned both questionnaires complete with consistent data. Of these, 44 received additional psychologic treatment by a psychologist, while 61 did not. Participation or non-participation in the psychologic treatment was the grouping variable in this study. RESULTS: For only 189/900 (21%) of the patients taking part in the study complete and consistent questionnaires were available for analysis. For the 105 orthopaedic patients, no significant score differences at arrival between the groups were detected (p = 0.38) using the Mann-Whitney U-test. At discharge, the mean rank sums for the psychological intervention group was 45.7, while it amounted to 58.3 in the control group, indicating significantly stronger improvement of symptoms in the intervention group (p = 0.04). CONCLUSIONS: Under routine conditions without any arrangements to ensure data quality a great amount of incomplete and/or inconsistent data has to be expected. For subsequent studies, this fact has to be taken into account. Irrespective of this aspect, complementary psychologic treatment of certain orthopaedic diseases seems to lead to significantly better results of treatment, as assessed by a psychophysiological questionnaire.


Subject(s)
Arousal/physiology , Musculoskeletal Diseases/rehabilitation , Patient Admission , Patient Care Team , Psychotherapy , Sick Role , Adult , Cognitive Behavioral Therapy , Disability Evaluation , Female , Humans , Male , Middle Aged , Musculoskeletal Diseases/physiopathology , Musculoskeletal Diseases/psychology , Outcome and Process Assessment, Health Care , Pilot Projects , Psychophysiology , Psychotherapy, Group
10.
Am J Physiol ; 274(2): H456-66, 1998 02.
Article in English | MEDLINE | ID: mdl-9486248

ABSTRACT

The effects of chronic treatment with growth hormone (porcine GH, 0.56 mg.kg-1.day-1 s.c.) were examined in dogs with heart failure induced by rapid ventricular pacing (240 beats/min) for 4 wk. Fourteen conscious dogs were studied 2-3 wk after surgical instrumentation with catheters in the descending aorta and left atrium, a pressure gauge in the left ventricle (LV), a flow probe around the ascending aorta, pacing leads on the ventricular free wall and left atrium, and ultrasonic crystals on the opposing anterior and posterior endomyocardium of the LV. GH treatment for 4 wk significantly increased both body weight and plasma insulin-like growth factor 1 (IGF-1) compared with vehicle-treated dogs (P < 0.01, +2.0 +/- 0.5 vs. +0.3 +/- 1.1 kg; 1,043 +/- 218 vs. 241 +/- 64 ng/ml, respectively). However, the changes in resting LV systolic (i.e., both isovolumic and ejection phases) and diastolic function (i.e., isovolumic relaxation time constant tau) and the systemic vascular resistance were similar for the GH- and vehicle-treated groups during the development of heart failure. LV contractile reserve, assessed with step infusion of isoproterenol or dobutamine challenge, was markedly attenuated after heart failure, but there were no differences between the GH- and vehicle-treated groups. During the progression of heart failure, the increases in plasma atrial natriuretic peptide correlated (P < 0.01) directly with left atrial pressure and inversely with LV circumferential fiber shortening. However, GH treatment did not substantially modify these relationships. In addition, renal function and myocardial ultrastructure at the advanced stage of heart failure also showed similar changes for the GH- and vehicle-treated groups. We conclude that in conscious dogs during the development of congestive heart failure produced by rapid ventricular pacing, GH at a dose that increases body weight and plasma IGF-1 levels does not affect LV performance or systemic vascular dynamics.


Subject(s)
Growth Hormone/pharmacology , Heart Failure/physiopathology , Adrenergic beta-Agonists/pharmacology , Animals , Atrial Natriuretic Factor/blood , Blood Pressure , Body Weight , Cardiac Pacing, Artificial , Dobutamine/pharmacology , Dogs , Heart Failure/etiology , Heart Failure/pathology , Insulin-Like Growth Factor I/metabolism , Isoproterenol/pharmacology , Kidney/physiopathology , Microscopy, Electron , Myocardial Contraction , Myocardium/ultrastructure , Vascular Resistance , Ventricular Function, Left
12.
J Cereb Blood Flow Metab ; 17(11): 1157-65, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9390647

ABSTRACT

In the cerebral circulation, endothelin-A receptor activation mediates marked prolonged vasoconstriction whereas endothelin-B (ETB) receptor activation effects dilation. In contrast to some peripheral vascular beds, ET(B) receptor-induced vasoconstriction has not yet been demonstrated in brain vessels. In this study in chloralose-anesthetized cats, with perivascular microapplications of ET(B) selective agonist (BQ-3020) and antagonist (BQ-788), we investigated whether ET(B) receptor-mediated constriction could be uncovered in cortical arterioles in vivo. In addition, we examined whether normal dilator response to ET(B) receptor activation is preserved in postischemic cerebral arterioles. The first microapplication of the selective ET(B) receptor agonist BQ-3020 (1 micromol/L) onto a pial cortical arteriole elicited marked dilation (caliber increased by 26.3 +/- 15.1% from preinjection baseline). A second application of BQ-3020 (10-minute interval) onto the same vessel failed to evoke any significant vasomotor response. Subsequent (third and fourth) adventitial microapplication of the ET(B) receptor agonist on the same arteriolar site effected a significant constriction of cerebral arterioles (-15.3 +/- 12.7% and -9.7 +/- 6.3% from preinjection baseline, respectively, at 20 and 30 minutes after the first application). The pial arterioles did not display tachyphylaxis to repeated applications of potassium (10 mmol/L). The perivascular application of the ET(B) receptor antagonist BQ-788 (0.001 to 1 micromol/L) had no effect on arteriolar caliber per se but blocked both BQ-3020-induced dilation (inhibitory concentration approximately 5 nmol/L) and vasoconstriction elicited by repeated activation of ET(B) receptors. After middle cerebral artery occlusion, most of the arterioles examined displayed a sustained dilation. The microapplication of BQ-3020 into the perivascular space surrounding postischemic dilated arterioles elicited a constriction of a similar magnitude to that induced by application of CSF (-17 +/- 7% and -17 +/- 7% from preinjection baseline, respectively). The adventitial microapplication of the ET(B) receptor antagonist (BQ-788, 0.1 micromol/L) on postocclusion dilated pial arterioles effected no change in the arteriolar caliber when compared with preinjection baseline. This BQ-788-induced response was significantly different from that induced by perivascular microinjection of CSF (P < 0.001, analysis of variance). These investigations indicate that (1) repeated activation of ET(B) receptors displays tachyphylaxis of the vasodilator response but also uncovers significant constriction of cerebral arterioles in vivo; (2) the ability of BQ-3020 to elicit dilation is lost within 30 minutes of induced focal ischemia; and (3) ET(B)-mediated contractile tone contributes in a small but significant manner in limiting postischemia dilation of cortical pial arterioles.


Subject(s)
Brain Ischemia/metabolism , Cerebrovascular Circulation/physiology , Receptors, Endothelin/metabolism , Animals , Arterioles/drug effects , Arterioles/metabolism , Brain Ischemia/physiopathology , Cats , Cerebrovascular Circulation/drug effects , Drug Combinations , Endothelins/pharmacology , Female , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Piperidines/pharmacology , Receptor, Endothelin B , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
13.
Eur J Pharmacol ; 294(2-3): 429-37, 1995 Dec 29.
Article in English | MEDLINE | ID: mdl-8750703

ABSTRACT

L-163,017 (6-[benzoylamino]-7-methyl-2-propyl-3-[[2'-(N-(3-methyl-1-butoxy) carbonylaminosulfonyl) [1,1']-biphenyl-4-yl]methyl]-3H-imidazo[4,5-b]pyridine) inhibited specific 125I-[Sar1, Ile8]angiotensin II binding to angiotensin AT1 receptor (Ki = 0.11-0.20 nM) in rabbit aorta, rat adrenal and human angiotensin AT1 receptor in CHO (Chinese hamster ovary transformed) cells and to AT2 receptor (Ki = 0.14-0.23 nM) in rat adrenal and brain receptors. L-163,017 also had a high affinity in the presence of bovine serum albumin (2 mg/ml), for angiotensin AT1 and AT2 receptors on human adrenal (Ki 3.9 and 4.3 nM), aorta (Ki 0.45 and 0.96 nM) and kidney (Ki 3.6 and 2.3 nM). The much higher Ki values in human tissues were likely due to the presence of bovine serum albumin in the binding assay buffer since L-163,017 had Ki values of 0.13 +/- 0.04 and 2.0 +/- 0.04 nM in the absence and presence of bovine serum albumin, respectively, in inhibiting 125I-[Sar1,Ile8]angiotensin II binding to angiotensin AT1 receptor in rat adrenal membranes. Scatchard analysis of 125I-[Sar1,Ile8]angiotensin II binding in the presence of bovine serum albumin (2 mg/ml) in rabbit aorta and bovine cerebellum indicated a competitive interaction of L-163,017 with angiotensin AT1 and AT2 receptors (Ki values 2.5 and 2.1 nM respectively). L-163,017 inhibited angiotensin II-induced aldosterone release in rat adrenal demonstrating that L-163,017 acted as a competitive antagonist (pA2 = 9.9) and lacked agonist activity. L-163,017 also inhibited angiotensin II responses in rat vascular tissues. The specificity of L-163,017 was shown by its lack of activity on the above functional responses produced by other agonists and in several binding assays.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin Receptor Antagonists , Imidazoles/pharmacology , Pyridines/pharmacology , Aldosterone/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , CHO Cells , Cattle , Cricetinae , Humans , Imidazoles/metabolism , In Vitro Techniques , Male , Pyridines/metabolism , Rabbits , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/metabolism , Vasoconstriction/drug effects
14.
Eur J Pharmacol ; 294(2-3): 439-50, 1995 Dec 29.
Article in English | MEDLINE | ID: mdl-8750704

ABSTRACT

L-163,017 (6-[benzoylamino]-7-methyl-2-propyl-3-[[2'-(N-(3-methyl-1-butoxy) carbonylaminosulfonyl)[1,1']-biphenyl-4-yl]methyl]-3H-imidazo[4,5- b]pyridine) is a potent, orally active, nonpeptide angiotensin II receptor antagonist. Conscious rats and dogs were dosed p.o. and i.v.; in both species the plasma bioequivalents are similar at the angiotensin AT1 and AT2 receptor sites indicating balanced activity is maintained in vivo. L-163,017 prevents the pressor response to intravenous (i.v.) angiotensin II in the conscious rat, dog, and rhesus monkey. L-163,017 also significantly reduces blood pressure in a renin-dependent model of hypertension, similar to an angiotensin converting enzyme inhibitor (Enalapril) and an angiotensin AT1 receptor-selective antagonist (L-159,282). These studies indicate that neither the angiotensin AT2 receptor nor bradykinin is important in the acute antihypertensive activity of angiotensin converting enzyme inhibitors or angiotensin II receptor antagonists.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin Receptor Antagonists , Imidazoles/pharmacology , Pyridines/pharmacology , Animals , Antihypertensive Agents/pharmacology , Biological Availability , Blood Pressure/drug effects , Dogs , Female , Imidazoles/metabolism , Macaca mulatta , Male , Pyridines/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/metabolism
15.
J Med Chem ; 38(19): 3741-58, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7562905

ABSTRACT

In order to block the effects induced by the interactions between angiotensin II (AII) and both AT1 and AT2 receptors, we have pursued the discovery of orally active non-peptide AII antagonists that exhibit potent and equal affinity for human AT1 and AT2 receptor subtypes. A series of previously prepared nanomolar (IC50) trisubstituted 1,2,4-triazolinone biphenyl-sulfonamide dual-acting AII antagonists has been modified at five different positions in order to increase AT2 binding affinity, maintain AT1 activity, and reduce the human adrenal AT2/AT1 potency ratio (IC50 ratio) from > or = 10. The targeted human adrenal potency ratio of < or = 1 was achieved with a number of compounds possessing an ethyl group at C5 of the triazolinone and a 3-fluoro substituent at the N4-biarylmethyl moiety. The most favored of these was compound 44 which exhibited subnanomolar potency at both the AT1 (rabbit aorta) and AT2 (rat midbrain) receptors, with a slight preference for the latter, and had a human adrenal AT2/AT1 IC50 ratio of 1. This tert-butyl sulfonylcarbamate with an N2-[2-bromo-5-(valerylamino)phenyl] substituent had excellent iv activity at 1 mg/kg (100% peak inhibition, > or = 4 h duration of action) and is orally active at 3 mg/kg with > 6 h duration of action in a conscious rat model. The present study shows that the NH of the amide on the N2-aryl moiety is not required for subnanomolar binding affinity to either receptor subtype, although a keto functionality at this position is essential for acceptable AT2 binding. Receptor-ligand binding interactions derived from the structure-activity relationships are discussed with respect to both receptor subtypes.


Subject(s)
Angiotensin Receptor Antagonists , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/pharmacology , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Triazoles/chemical synthesis , Triazoles/pharmacology , Administration, Oral , Adrenal Glands/metabolism , Angiotensin II/antagonists & inhibitors , Angiotensin II/metabolism , Animals , Biphenyl Compounds/chemistry , Biphenyl Compounds/metabolism , Blood Pressure/drug effects , Humans , Mesencephalon/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/metabolism , Triazoles/chemistry , Triazoles/metabolism
16.
Clin Exp Hypertens ; 17(6): 931-53, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7581262

ABSTRACT

The present study was designed to characterize the in vivo pharmacology of L-159,913 (4-[[2'-(N-benzoylsulfamoyl)biphenyl-4-yl]-5butyl-2,4-dihydr o-2- [2-(trifluoromethyl)phenyl]-3H-1,2,4-triazol-3-one); a potent All receptor antagonist. In normotensive rats, dogs, rhesus monkeys, and chimpanzees, L-159,913 inhibited All-induced elevations in blood pressure. In conscious rats, the relative potencies (ED50) were 0.51 mg/kg i.v. and 0.72 mg/kg p.o. Duration of action with single i.v. or p.o. doses exceeded 6 hr in rats. L-159,913 was 3 times less potent than losartan in rats and equipotent to losartan in monkeys. All induced elevation of plasma aldosterone in rats was also inhibited by L-159,913. L-159,913 was antihypertensive in high renin hypertensive rats (aortic coarctation). The maximum hypotensive response to an acute dose of L-159,913 (10 mg/kg, po) was equal to that of enalaprilat (0.3 mg/kg, iv) in this renin dependent animal model. In conscious normotensive dogs, L-159,913 had a moderate diuretic, natriuretic and kaliuretic response with no effect on glomerular filtration rate, effective renal plasma flow or filtration fraction, suggesting a tubular site of action. L-159,913 is a selective and potent All receptor antagonist with good oral activity, long duration of action and antihypertensive efficacy.


Subject(s)
Angiotensin Receptor Antagonists , Antihypertensive Agents/pharmacology , Triazoles/pharmacology , Aldosterone/blood , Angiotensin II/pharmacology , Animals , Biphenyl Compounds/pharmacology , Blood Pressure/drug effects , Dogs , Female , Hypertension/drug therapy , Hypertension/physiopathology , Imidazoles/pharmacology , Kidney/drug effects , Kidney/physiology , Losartan , Macaca mulatta , Male , Natriuresis/drug effects , Pan troglodytes , Potassium/urine , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/drug effects , Receptors, Angiotensin/physiology , Renin/physiology , Tetrazoles/pharmacology
17.
J Hypertens Suppl ; 13(1): S15-21, 1995 Jul.
Article in English | MEDLINE | ID: mdl-18800451

ABSTRACT

BACKGROUND: Clinical experience with angiotensin converting enzyme (ACE) inhibitors has shown that inhibition of the renin-angiotensin system is effective therapy for hypertension and heart failure. Losartan (DuP753, MK954, cozaar) is the first non-peptidic drug that inhibits the renin-angiotensin system by selectively blocking the interaction of angiotensin II with its receptor. DIFFERENCES BETWEEN LOSARTAN AND ACE INHIBITORS: Pharmacological differences between ACE inhibitors and losartan could affect comparative efficacy and/or safety. In addition to angiotensin I, ACE has other substrates (e.g. kinins). Blocking the metabolism of kinins with ACE inhibitors could be beneficial (e.g. vasodilation) and/or elicit side effects (e.g. cough) which will not be produced by losartan. Non-ACE pathways of angiotensin II formation have been described (e.g. angiotensin I convertase) which may limit the ability of ACE inhibitors to prevent formation of angiotensin II in all tissues. Losartan blocks angiotensin II responses irrespective of the route or site of angiotensin II formation. Two binding sites for angiotensin II are widely accepted, AT1 and AT2. Losartan blocks only AT1 sites while ACE inhibitors functionally block angiotensin II interaction with both sites. Since the physiological role for AT2 sites is unknown, the relevance of this difference between ACE inhibitors and losartan is questionable. HYPERTENSION: In animal models of hypertension, the efficacy of losartan is equivalent to the efficacy of ACE inhibitors. In animal models that reflect complications of hypertension, such as kidney dysfunction, cardiac and vascular hypertrophy and stroke, losartan and ACE inhibitors are also equally effective. From these results, kinin potentiation and lack of inhibition of angiotensin I convertase do not lead to differences in pharmacological efficacy between ACE inhibitors and losartan. Therefore, with respect to therapeutic efficacy, results in animal models indicate that losartan will display the beneficial pharmacology of ACE inhibitors without the detrimental side effects attributed to kinin potentiation.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Blood Pressure/drug effects , Hypertension/physiopathology , Losartan/pharmacology , Angiotensin II Type 1 Receptor Blockers/adverse effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Blood Pressure/physiology , Cardiomegaly/prevention & control , Disease Models, Animal , Kidney Diseases/prevention & control , Losartan/adverse effects , Rats , Rats, Inbred Dahl , Rats, Inbred SHR , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology
18.
J Pharmacol Exp Ther ; 272(2): 612-8, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7853174

ABSTRACT

The affinities of 13 angiotensin II antagonists for the AT1 subtype determined in vitro with tissue homogenates were shown not to correlate well with in vivo pharmacologic potency. The addition of human serum albumin to the in vitro assay to mimic in vivo plasma protein interactions reduced the measured affinity by reducing the effective free concentrations of antagonists, but the resulting affinities were not predictive of the in vivo effects. Using an in vivo radioligand competition assay, in which receptor occupancy is demonstrated via competitive blockade of the in vivo binding of [125I][Sar1,Ile8]angiotensin II to AT1 receptors in rat kidney cortex, we demonstrated that the in vivo pharmacologic potencies reflect receptor occupancy. By comparing the effects of rat plasma and bovine serum albumin on the in vitro affinity of two antagonists, we suggest that the use of plasma would alter free plasma concentrations in a manner more consistent with in vivo measures of potencies.


Subject(s)
Angiotensin II/antagonists & inhibitors , Receptors, Angiotensin/metabolism , 1-Sarcosine-8-Isoleucine Angiotensin II/metabolism , Angiotensin Receptor Antagonists , Animals , Binding, Competitive , Imidazoles/metabolism , Imidazoles/pharmacology , Male , Protein Binding , Rats , Rats, Sprague-Dawley , Serum Albumin/metabolism , Tetrazoles/metabolism , Tetrazoles/pharmacology
19.
J Med Chem ; 37(26): 4464-78, 1994 Dec 23.
Article in English | MEDLINE | ID: mdl-7799397

ABSTRACT

Angiotensin II (AII), the endogenous peptide ligand of the AII receptor, has equivalent high affinity for both the AT1 and AT2 receptor subtypes while most of the reported nonpeptide AII antagonists are AT1-selective. In an effort to identify dual AT1/AT2 nonpeptide AII antagonists, we have pursued modifications of previously prepared trisubstituted 1,2,4-triazolinone biphenylsulfonamides which exhibited subnanomolar in vitro AT1 (rabbit aorta) AII antagonism and AT2 (rat midbrain) IC50 values of < 40 nM. Present results show that a suitable amide (or reversed amide) side chain appropriately positioned on the N2-aryl group of these compounds gave > 15-fold enhancement in AT2 binding affinity without sacrificing nanomolar AT1 potency (IC50). This added amide, combined with an appropriate choice of the N-substituent on the sulfonamide and the ortho substituent on the N2-aryl group, led to an analogue (46, L-163,-007) which exhibited subnanomolar AT1 binding affinity and an AT2/AT1 IC50 ratio of 3. This compound showed excellent iv activity at 1 mg/kg and oral efficacy at 3 mg/kg with > 6 h duration in a conscious rat model. Available data suggest that the newly introduced amide side chain, mandatory for low nanomolar binding affinity at the AT2 receptor, is well-tolerated by the AT1 receptor and has minimal effect on the in vivo properties of these molecules.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin Receptor Antagonists , Sulfonamides/chemical synthesis , Angiotensin II/metabolism , Animals , Blood Pressure/drug effects , In Vitro Techniques , Male , Rabbits , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfonamides/pharmacology
20.
Lab Invest ; 71(4): 543-51, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7967510

ABSTRACT

BACKGROUND: Juxtaglomerular (JG) cell hypertrophy and hyperplasia were investigated in rhesus monkeys given angiotensin II (AII) AT1 receptor antagonists L-158,338 and DUP 753 (MK-0954, losartan). EXPERIMENTAL DESIGN: In 2 initial studies, L-158,338 was given orally at 10, 30, and 90 mg/kg/day for 3 or 14 weeks. To investigate the observed JG hypertrophy and hyperplasia, in a third 5-week experiment L-158,338 was given alone at 90 mg/kg/day, or with physiologic saline supplementation at 25 ml/kg/day, or coadministered with the angiotensin converting enzyme inhibitor enalapril at 10 mg/kg/day. Physiologic saline was given to attempt to suppress renin release through volume expansion and/or sodium retention. Enalapril was given to lower plasma AII levels and observe whether JG cell hypertrophy and hyperplasia were increased or decreased. For comparison, DUP 753 was given at 90 and 300 mg/kg/day. Plasma renin activity and AII concentration were measured in this study. RESULTS: Dose- and time-dependent increases in JG cell hypertrophy and hyperplasia were seen in the 2 initial experiment. In the third experiment, plasma renin activity and AII concentration were increased 3-fold and 6-fold over pretest values by L-158,338 at 90 mg/kg/day for 5 weeks. Saline supplementation had no effect on these parameters but diminished the group mean severity grade for JG hypertrophy and hyperplasia from 1.5 to 1.0. Enalapril coadministration had no effect on plasma renin activity, whereas it blunted the plasma AII increase caused by L-158,338 and increased the group mean grade to 2.5. DUP 753 at 300 mg/kg/day produced similar increases in plasma renin activity and AII concentration but only resulted in grade 1 JG cell hypertrophy and hyperplasia. CONCLUSIONS: L-158,338-induced JG cell hypertrophy and hyperplasia is an exaggerated pharmacologic response that can be modulated by saline supplementation and angiotensin converting enzyme inhibition. These results suggest that decreased renal perfusion or altered sodium homeostasis and plasma AII concentration are important variables that contribute to AT1 receptor blockade to induce JG cell hypertrophy and hyperplasia.


Subject(s)
Angiotensin Receptor Antagonists , Biphenyl Compounds/pharmacology , Imidazoles/pharmacology , Immunosuppressive Agents/pharmacology , Juxtaglomerular Apparatus/pathology , Pyridines/pharmacology , Tetrazoles/pharmacology , Angiotensin II/blood , Animals , Biphenyl Compounds/adverse effects , Dose-Response Relationship, Drug , Enalapril/pharmacology , Hyperplasia/blood , Hyperplasia/chemically induced , Hyperplasia/pathology , Hypertrophy/blood , Hypertrophy/chemically induced , Hypertrophy/pathology , Imidazoles/adverse effects , Immunosuppressive Agents/adverse effects , Juxtaglomerular Apparatus/drug effects , Juxtaglomerular Apparatus/metabolism , Losartan , Macaca mulatta , Pyridines/adverse effects , Renin/blood , Tetrazoles/adverse effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...