Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(5): 4100-4119, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38482828

ABSTRACT

C5a is an anaphylatoxin protein produced by the cleavage of the complement system's component C5 protein. It signals through the G-protein-coupled receptor C5a receptor 1 (C5aR1) to induce the chemotaxis of primarily neutrophils and monocytes and the release of inflammatory molecules. A large body of evidence linking C5aR1 signaling to acute and chronic inflammatory disorders has triggered interest in developing potent C5aR antagonists. Herein we report the discovery of new C5aR1 antagonistic chemical classes. Many representatives showed low nanomolar IC50 values in a C5aR1 ß-arrestin-2 recruitment assay, inhibiting the migration of human neutrophils toward C5a and the internalization of the receptor in human whole blood. Two leading compounds were characterized further in vivo. Target engagement of the receptor by these two C5aR1 antagonists was demonstrated in vivo. In particular, the inhibition of migration in vitro with the two compounds further translated in a dose-dependent efficacy in a rat model of C5a-induced neutrophilia.


Subject(s)
Complement C5a , Receptor, Anaphylatoxin C5a , Humans , Rats , Animals , Complement C5a/metabolism , Chemotaxis , Monocytes/metabolism , Neutrophils/metabolism
2.
J Med Chem ; 67(4): 2379-2396, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38349223

ABSTRACT

Lysophosphatidic acid receptor 1 (LPAR1) antagonists show promise as potentially novel antifibrotic treatments. In a human LPAR1 ß-arrestin recruitment-based high-throughput screening campaign, we identified urea 19 as a hit with a LPAR1 IC50 value of 5.0 µM. Hit-to-lead activities revealed that one of the urea nitrogen atoms can be replaced by carbon and establish the corresponding phenylacetic amide as a lead structure for further optimization. Medicinal chemistry efforts led to the discovery of piperidine 18 as a potent and selective LPAR1 antagonist with oral activity in a mouse model of LPA-induced skin vascular leakage. The molecular scaffold of 18 shares no obvious structural similarity with any other LPAR1 antagonist disclosed so far.


Subject(s)
Amides , Receptors, Lysophosphatidic Acid , Mice , Animals , Humans , Disease Models, Animal , Urea
3.
J Med Chem ; 67(4): 2397-2424, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38349250

ABSTRACT

Piperidine 3 is a potent and selective lysophosphatidic acid receptor subtype 1 receptor (LPAR1) antagonist that has shown efficacy in a skin vascular leakage target engagement model in mice. However, compound 3 has very high human plasma protein binding and high clearance in rats, which could significantly hamper its clinical development. Continued lead optimization led to the potent, less protein bound, metabolically stable, and orally active azetidine 17. Rat pharmacokinetics (PK) studies revealed that 17 accumulated in the liver. In vitro studies indicated that 17 is an organic anion co-transporting polypeptide 1B1 (OATP1B1) substrate. Although analogue 24 was no longer a substrate of OATP1B1, PK studies suggested that the compound undergoes enterohepatic recirculation. Replacing the carboxylic acidic side chain by a non-acidic sulfamide moiety and further fine-tuning of the scaffold yielded the potent, orally active LPAR1 antagonist 49, which was selected for preclinical development for the treatment of fibrotic diseases.


Subject(s)
Organic Anion Transporters , Receptors, Lysophosphatidic Acid , Humans , Rats , Mice , Animals , Receptors, Lysophosphatidic Acid/metabolism , Liver/metabolism
4.
Clin Transl Sci ; 16(7): 1220-1231, 2023 07.
Article in English | MEDLINE | ID: mdl-37042126

ABSTRACT

ACT-1014-6470 is an orally available complement factor 5a receptor 1 antagonist and a novel treatment option in auto-inflammatory diseases. The in vitro inhibition potential of ACT-1014-6470 on cytochrome P450 isozymes (CYPs) and its effect on the pharmacokinetics (PK) of the CYP2C19 and CYP3A4 substrates omeprazole and midazolam, respectively, in humans were assessed. In vitro assays were conducted with isoform-specific substrates in human liver microsomes. In an open-label, two-period, fixed-sequence cocktail study, single doses of 20 mg omeprazole and 2 mg midazolam were administered concomitantly to 20 healthy male subjects alone (treatment A) and after a single dose of 100 mg ACT-1014-6470 (treatment B) under fed conditions. Safety and PK assessments were performed. Geometric mean ratios (GMRs) and 90% confidence intervals (CIs) of noncompartmental PK parameters of treatment B versus treatment A were calculated. In vitro, no time-dependent inhibition was observed and the lowest inhibition constant of 4.3 µM ACT-1014-6470 was recorded for CYP2C19. In humans, GMRs (90% CI) of omeprazole PK were 1.9 (1.5-2.5) for maximum plasma concentration (Cmax ) and 1.9 (1.5-2.3) for area under the plasma concentration-time curve from 0 to 12 h (AUC0-12 h ). Midazolam PK showed GMRs (90% CI) of 1.1 (1.1-1.2) for Cmax and 1.5 (1.4-1.6) for AUC0-24 h . All treatments were well-tolerated. In line with in vitro results and regulatory risk factor calculation, the increased exposure to omeprazole and midazolam in humans after concomitant administration with a single dose of 100 mg ACT-1014-6470 reflected a weak inhibition of CYP2C19 and CYP3A4.


Subject(s)
Cytochrome P-450 CYP3A , Factor Va , Humans , Male , Midazolam/pharmacokinetics , Cytochrome P-450 CYP2C19 , Drug Interactions , Cytochrome P-450 Enzyme System , Omeprazole/pharmacokinetics
5.
ChemMedChem ; 18(10): e202300030, 2023 05 16.
Article in English | MEDLINE | ID: mdl-36892179

ABSTRACT

The dual orexin receptor antagonist daridorexant was approved in 2022 in the USA and EU for the treatment of insomnia. The purpose of this study was the identification of its metabolic pathways and the human cytochrome P450 (P450) enzymes involved in its biotransformation. With human liver microsomes, daridorexant underwent hydroxylation at the methyl group of the benzimidazole moiety, oxidative O-demethylation of the anisole to the corresponding phenol, and hydroxylation to a 4-hydroxy piperidinol derivative. While the chemical structures of the benzylic alcohol and the phenol proved to be products of standard P450 reactions, 1D and 2D NMR data of the latter hydroxylation product was incompatible with the initially postulated hydroxylation of the pyrrolidine ring and suggested the disappearance of the pyrrolidine ring and formation of a new 6-membered ring. Its formation is best explained by initial hydroxylation of the pyrrolidine ring in 5-position to yield a cyclic hemiaminal. Hydrolytic ring opening then results in an aldehyde that subsequently cyclizes onto one of the benzimidazole nitrogen atoms to yield the final 4-hydroxy piperidinol. The proposed mechanism was substantiated using an N-methylated analogue, which might hydrolyze to the open-chain aldehyde but cannot undergo the final cyclization step. CYP3A4 was the major P450 enzyme responsible for daridorexant metabolism, accounting for 89 % of metabolic turnover.


Subject(s)
Cytochrome P-450 CYP3A , Orexin Receptor Antagonists , Humans , Cytochrome P-450 CYP3A/metabolism , Orexin Receptor Antagonists/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Pyrrolidines/pharmacology , Microsomes, Liver/metabolism , Benzimidazoles/pharmacology , Phenols/pharmacology
6.
Xenobiotica ; 53(3): 173-183, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36809238

ABSTRACT

Daridorexant is a dual orexin receptor antagonist developed for the treatment of insomnia disorder and has shown improvement in sleep outcomes and daytime functioning. The present work describes its biotransformation pathways in vitro and in vivo and provides a cross-species comparison between the animal species used in preclinical safety assessments and man.Daridorexant clearance is driven by metabolism along seven distinct pathways. Metabolic profiles were characterised by downstream products while primary metabolic products were of minor importance. The metabolic pattern differed between rodent species, with the rat reflecting the human pattern better than the mouse.In rodents, daridorexant is mostly excreted via the bile after extensive metabolism while urinary excretion was negligible in the rat. Only traces of the parent drug were detected in urine, bile, or faeces.Daridorexant has three major metabolites which are well covered in these preclinical safety species. All of them retain some residual affinity towards orexin receptors. However, none of these is considered to contribute to the pharmacological effect of daridorexant as their active concentrations in the human brain are too low.


Subject(s)
Orexin Receptor Antagonists , Sleep Initiation and Maintenance Disorders , Male , Rats , Humans , Mice , Animals , Imidazoles , Sleep Initiation and Maintenance Disorders/drug therapy , Pyrrolidines
7.
Clin Transl Sci ; 15(8): 1906-1915, 2022 08.
Article in English | MEDLINE | ID: mdl-35583936

ABSTRACT

Selatogrel is a potent and selective reversible P2Y12 receptor antagonist in development for early treatment of acute myocardial infarction via subcutaneous (s.c.) self-injection. Selatogrel is almost exclusively eliminated via the hepatobiliary route. Hepatic impairment is associated with reduced drug clearance and primary hemostasis. This single-center, open-label study investigated the effect of mild and moderate hepatic impairment on pharmacokinetics (PK) and pharmacodynamics (PD) of a single s.c. dose of selatogrel (16 mg). The study included groups of eight subjects with mild and moderate hepatic impairment, and matched healthy control subjects. Compared to healthy subjects, exposure to selatogrel in subjects with mild and moderate hepatic impairment was 30% and 108% (maximum plasma concentration [Cmax ]) and 47% and 212% (area under the concentration-time curve from zero to infinity [AUC0-∞ ]) higher, respectively. Hepatic impairment was associated with lower clearance and volume of distribution, whereas plasma protein binding was not affected. Marked inhibition of platelet aggregation (IPA > 80%) was attained within 30 min in all subjects and hepatic impairment prolonged IPA duration. Area under the effect curve was 60% and 160% higher in subjects with mild and moderate hepatic impairment, respectively. PK/PD modeling identified a change in the relationship between exposure and IPA, with a steeper concentration-effect relationship in healthy subjects compared to subjects with hepatic impairment. The combination of higher exposure and lower half-maximum inhibitory concentration resulted in longer lasting effect. In conclusion, hepatic impairment alters the PK/PD relationship leading to prolonged effects. Therefore, dose adjustments may be warranted in subjects with moderate hepatic impairment.


Subject(s)
Liver Diseases , Purinergic P2Y Receptor Antagonists , Area Under Curve , Humans , Liver Diseases/drug therapy , Organophosphonates , Pyrimidines
8.
Clin Pharmacokinet ; 60(10): 1349-1360, 2021 10.
Article in English | MEDLINE | ID: mdl-34002356

ABSTRACT

BACKGROUND AND OBJECTIVE: Daridorexant is a dual orexin receptor antagonist in clinical development for insomnia. As daridorexant is cleared mainly via cytochrome P450 (CYP) 3A4, the effect of hepatic impairment on the pharmacokinetics (PK), metabolism, and tolerability of daridorexant was evaluated. Sleep disorders are common in patients with liver cirrhosis and, therefore, sleep-promoting drugs with a better tolerability than currently available would be preferable, a premise that dual orexin receptor antagonists may fulfill. METHODS: This was a single-dose, open-label, phase I study. Subjects with mild (Child-Pugh A, N = 8) or moderate (Child-Pugh B, N = 8) liver cirrhosis and matched healthy control subjects (N = 8) received 25 mg of daridorexant orally. Blood samples were collected for 72 h post-dose for PK assessments of daridorexant and three major metabolites. RESULTS: Compared with healthy subjects, patients showed a decrease in total daridorexant area under the plasma concentration-time curve from zero to infinity (AUC0-inf) and maximum plasma concentration with a geometric mean ratio (GMR, 90% confidence interval [CI]) of 0.51 (0.28-0.92) and 0.50 (0.35-0.72) in Child-Pugh A and 0.74 (0.39-1.41) and 0.42 (0.29-0.60) in Child-Pugh B patients, respectively. Furthermore, the median time to reach maximum plasma concentration was slightly delayed (1.0 h [90% CI 0.0-2.0] in Child-Pugh A patients and 0.5 h [90% CI 0.0-1.5] in Child-Pugh B patients), while for Child-Pugh B patients, a doubling in half-life was observed (GMR [90% CI]: 2.09 [1.32-3.30]). Considering the high plasma protein binding (> 99%) and a 1.9-fold to 2.3-fold increase in the unbound fraction in patients, the PK of unbound daridorexant was also assessed. Compared with healthy subjects, Child-Pugh B patients had a higher AUC0-inf (GMR [90% CI] 1.60 [0.93-2.73]), a lower apparent plasma clearance (GMR [90% CI] 0.63 [0.37-1.07]), and the same doubling in the half-life observed for total daridorexant, whereas maximum plasma concentration and apparent volume of distribution were not different. Unbound daridorexant PK in Child-Pugh A patients did not differ from healthy subjects. In addition, the metabolic ratios (parent to metabolite), i.e., a marker of CYP 3A4 activity, of the two most abundant daridorexant metabolites were higher in patients with liver cirrhosis compared with healthy subjects. All treatment-emergent adverse events were transient and of mild or moderate intensity and no other treatment-related effects were apparent. CONCLUSIONS: No safety issue of concern was detected following administration of 25 mg of daridorexant in the study population. Moderate liver cirrhosis causes impaired hepatic clearance of unbound daridorexant, which prolongs the half-life. A 25-mg dose of daridorexant should, therefore, not be exceeded in Child-Pugh B patients. A dose adjustment is not required in Child-Pugh A patients, while avoidance of daridorexant in patients with Child-Pugh C cirrhosis is recommended. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov ID: NCT03713242.


Subject(s)
Liver Diseases , Orexin Receptor Antagonists , Area Under Curve , Humans , Imidazoles , Liver Cirrhosis/drug therapy , Pyrrolidines
9.
Pharmacol Res Perspect ; 8(4): e00619, 2020 08.
Article in English | MEDLINE | ID: mdl-32613761

ABSTRACT

The dual endothelin receptor antagonist macitentan was approved in 2013 for the treatment of pulmonary arterial hypertension. Macitentan is an inducer of cytochrome P450 expression in vivo in animal species but not in man. In rat and dog, changes in P450 expression manifest as autoinduction upon repeat dosing. The induction pattern, however, significantly differed between both species, and between male and female rats. While macitentan exposure steadily declined with dose in the dog, P450 induction was saturable in the rat reaching levels of 40%-60% and 60%-80% at steady-state in male and female animals, respectively. The nature and number of P450 enzymes involved in macitentan clearance were identified as a major reason for the observed species differences. In the dog, macitentan was metabolized by a single P450 enzyme, that is, Cyp3a12, whereas several members of the Cyp2c and Cyp3a families were involved in the rat. Macitentan selectively upregulated Cyp3a expression in rat, whereas the expression of the Cyp2c enzymes involved in macitentan metabolism remained mostly unchanged, eventually leading to a higher contribution of Cyp3a upon induction. Macitentan also induced CYP3A4 expression in human hepatocytes via initial activation of the human pregnane X receptor. No such induction was evident in humans at the therapeutic macitentan dose of 10 mg as shown in a clinical drug-drug interaction study with the CYP3A4 substrate sildenafil.


Subject(s)
Cytochrome P-450 Enzyme Inducers/pharmacology , Endothelin Receptor Antagonists/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Cytochrome P-450 Enzyme Inducers/administration & dosage , Dogs , Dose-Response Relationship, Drug , Drug Interactions , Endothelin Receptor Antagonists/administration & dosage , Enzyme Induction/drug effects , Female , Hepatocytes/metabolism , Humans , Male , Pregnane X Receptor/drug effects , Pregnane X Receptor/metabolism , Pulmonary Arterial Hypertension/drug therapy , Pyrimidines/administration & dosage , Rats , Rats, Sprague-Dawley , Species Specificity , Sulfonamides/administration & dosage
10.
J Pharmacol Exp Ther ; 368(3): 462-473, 2019 03.
Article in English | MEDLINE | ID: mdl-30622171

ABSTRACT

The endothelin (ET) system has emerged as a novel target for hypertension treatment where a medical need persists despite availability of several pharmacological classes, including renin angiotensin system (RAS) blockers. ET receptor antagonism has demonstrated efficacy in preclinical models of hypertension, especially under low-renin conditions and in hypertensive patients. We investigated the pharmacology of aprocitentan (N-[5-(4-bromophenyl)-6-[2-[(5-bromo-2-pyrimidinyl)oxy]ethoxy]-4-pyrimidinyl]-sulfamide), a potent dual ETA/ETB receptor antagonist, on blood pressure (BP) in two models of experimental hypertension: deoxycorticosterone acetate (DOCA)-salt rats (low-renin model) and spontaneously hypertensive rats [(SHR), normal renin model]. We also compared the effect of its combination with RAS blockers (valsartan and enalapril) with that of the combination of the mineraloreceptor antagonist spironolactone with the same RAS blockers on BP and renal function in hypertensive rats. Aprocitentan was more potent and efficacious in lowering BP in conscious DOCA-salt rats than in SHRs. In DOCA-salt rats, single oral doses of aprocitentan induced a dose-dependent and long-lasting BP decrease and 4-week administration of aprocitentan dose dependently decreased BP (statistically significant) and renal vascular resistance, and reduced left ventricle hypertrophy (nonsignificant). Aprocitentan was synergistic with valsartan and enalapril in decreasing BP in DOCA-salt rats and SHRs while spironolactone demonstrated additive effects with these RAS blockers. In hypertensive rats under sodium restriction and enalapril, addition of aprocitentan further decreased BP without causing renal impairment, in contrast to spironolactone. In conclusion, ETA/ETB receptor antagonism represents a promising therapeutic approach to hypertension, especially with low-renin characteristics, and could be used in combination with RAS blockers, without increasing the risk of renal impairment.


Subject(s)
Antihypertensive Agents/administration & dosage , Endothelin Receptor Antagonists/administration & dosage , Hypertension/drug therapy , Hypertension/physiopathology , Pyrimidines/administration & dosage , Renin-Angiotensin System/drug effects , Sulfonamides/administration & dosage , Animals , Antihypertensive Agents/pharmacology , Desoxycorticosterone Acetate/toxicity , Drug Therapy, Combination , Endothelin Receptor Antagonists/pharmacology , Hypertension/chemically induced , Male , Pyrimidines/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar , Renin-Angiotensin System/physiology , Sulfonamides/pharmacology
11.
Xenobiotica ; 48(7): 704-719, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28737453

ABSTRACT

1. The metabolism of selexipag has been studied in vivo in man and the main excreted metabolites were identified. Also, metabolites circulating in human plasma have been structurally identified and quantified. 2. The main metabolic pathway of selexipag in man is the formation of the active metabolite ACT-333679. Other metabolic pathways include oxidation and dealkylation reactions. All primary metabolites undergo subsequent hydrolysis of the sulphonamide moiety to their corresponding acids. ACT-333679 undergoes conjugation with glucuronic acid and aromatic hydroxylation to P10, the main metabolite detected in human faeces. 3. The formation of the active metabolite ACT-333679 is catalysed by carboxylesterases, while the oxidation and dealkylation reactions are metabolized by CYP2C8 and CYP3A4. CYP2C8 is the only P450 isoform catalysing the aromatic hydroxylation to P10. CYP2C8 together with CYP3A4 are also involved in the formation of several minor ACT-333679 metabolites. UGT1A3 and UGT2B7 catalyse the glucuronidation of ACT-333679. 4. The potential of selexipag to inhibit or induce cytochrome P450 enzymes or drug transport proteins was studied in vitro. Selexipag is an inhibitor of CYP2C8 and CYP2C9 and induces CYP3A4 and CYP2C9 in vitro. Also, selexipag inhibits the transporters OATP1B1, OATP1B3, OAT1, OAT3, and BCRP. However, due to its low dose and relatively low unbound exposure, selexipag has a low potential for causing drug-drug interactions.


Subject(s)
Acetamides/metabolism , Acetamides/pharmacology , Pyrazines/metabolism , Pyrazines/pharmacology , Receptors, Epoprostenol/agonists , Acetamides/blood , Acetamides/chemistry , Acetates/pharmacology , Drug Interactions , Enzyme Inhibitors/pharmacology , Esterases/antagonists & inhibitors , Esterases/metabolism , Hepatocytes/metabolism , Humans , Membrane Transport Proteins/metabolism , Metabolic Networks and Pathways/drug effects , Metabolome , Metabolomics , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , NADP/metabolism , Pyrazines/blood , Pyrazines/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Epoprostenol/metabolism , Recombinant Proteins/metabolism
12.
ChemMedChem ; 11(18): 1995-2014, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27471138

ABSTRACT

More than 40 % of the world's population is at risk of being infected with malaria. Most malaria cases occur in the countries of sub-Saharan Africa, Central and South America, and Asia. Resistance to standard therapy, including artemisinin combinations, is increasing. There is an urgent need for novel antimalarials with new mechanisms of action. In a phenotypic screen, we identified a series of phenylalanine-based compounds that exhibit antimalarial activity via a new and yet unknown mechanism of action. Our optimization efforts culminated in the selection of ACT-451840 [(S,E)-N-(4-(4-acetylpiperazin-1-yl)benzyl)-3-(4-(tert-butyl)phenyl)-N-(1-(4-(4-cyanobenzyl)piperazin-1-yl)-1-oxo-3-phenylpropan-2-yl)acrylamide] for clinical development. Herein we describe our optimization efforts from the screening hit to the potential drug candidate with respect to antiparasitic activity, drug metabolism and pharmacokinetics (DMPK) properties, and in vivo pharmacological efficacy.


Subject(s)
Acrylamides/pharmacology , Antimalarials/pharmacology , Drug Discovery , Malaria/drug therapy , Piperazines/pharmacology , Plasmodium falciparum/drug effects , Acrylamides/chemical synthesis , Acrylamides/chemistry , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Molecular Structure , Parasitic Sensitivity Tests , Piperazines/chemical synthesis , Piperazines/chemistry , Structure-Activity Relationship
13.
Clin Pharmacokinet ; 55(3): 369-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26385839

ABSTRACT

INTRODUCTION: Macitentan is a novel dual endothelin receptor antagonist for the treatment of pulmonary arterial hypertension (PAH). It is metabolized by cytochrome P450 (CYP) enzymes, mainly CYP3A4, to its active metabolite ACT-132577. METHODS: A physiological-based pharmacokinetic (PBPK) model was developed by combining observations from clinical studies and physicochemical parameters as well as absorption, distribution, metabolism and excretion parameters determined in vitro. RESULTS: The model predicted the observed pharmacokinetics of macitentan and its active metabolite ACT-132577 after single and multiple dosing. It performed well in recovering the observed effect of the CYP3A4 inhibitors ketoconazole and cyclosporine, and the CYP3A4 inducer rifampicin, as well as in predicting interactions with S-warfarin and sildenafil. The model was robust enough to allow prospective predictions of macitentan-drug combinations not studied, including an alternative dosing regimen of ketoconazole and nine other CYP3A4-interacting drugs. Among these were the HIV drugs ritonavir and saquinavir, which were included because HIV infection is a known risk factor for the development of PAH. CONCLUSION: This example of the application of PBPK modeling to predict drug-drug interactions was used to support the labeling of macitentan (Opsumit).


Subject(s)
Endothelin A Receptor Antagonists/pharmacokinetics , Endothelin B Receptor Antagonists/pharmacokinetics , Models, Biological , Pyrimidines/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Cyclosporine/pharmacology , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions , Humans , Ketoconazole/pharmacology , Male , Pyrimidines/blood , Rifampin/pharmacology , Sildenafil Citrate/pharmacology , Sulfonamides/blood , Warfarin/pharmacology
14.
J Cardiovasc Pharmacol ; 66(5): 457-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26230396

ABSTRACT

AIMS: We compared the efficacy of macitentan, a novel dual endothelin A/endothelin B receptor antagonist, with that of another dual endothelin receptor antagonist, bosentan, in a rat model of non-vasoreactive pulmonary hypertension (PH) with particular emphasis on right ventricular (RV) remodeling. METHODS AND RESULTS: Unlike monocrotaline or hypoxic/sugen rats, bleomycin-treated rats presented a non-vasoreactive PH characterized by the absence of pulmonary dilatation to adenosine. We therefore chose the bleomycin rat model to compare the effects of the maximally effective doses of macitentan and bosentan on pulmonary vascular and RV remodeling. Macitentan (100 mg·kg(-1)·d(-1)), but not bosentan (300 mg·kg(-1)·d(-1)), significantly prevented pulmonary vascular remodeling, RV hypertrophy, and cardiomyocyte diameter increase. Cardiac protection by macitentan was associated with a significant attenuation of genes related to cell hypertrophy and extracellular matrix remodeling. Microautoradiography and high performance liquid chromatography analysis showed greater distribution of macitentan than bosentan in the RV and pulmonary tissue. CONCLUSIONS: Macitentan was more efficacious than bosentan in preventing the development of pulmonary and RV hypertrophies in a model of non-vasoreactive PH. Greater ability to distribute into the tissue could contribute to the greater structural improvement by macitentan compared with bosentan.


Subject(s)
Endothelin Receptor Antagonists/pharmacology , Heart Ventricles/drug effects , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/prevention & control , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects , Animals , Bleomycin , Bosentan , Disease Models, Animal , Gene Expression Regulation , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Male , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats, Wistar , Time Factors , Vascular Remodeling/drug effects
15.
Bioorg Med Chem Lett ; 25(18): 3941-6, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26231163

ABSTRACT

Chemical evolution of mibefradil resulted in the identification of novel bridged tetrahydronaphthalene derivatives as potent T/L-type calcium channel blockers. A SAR study, in vitro and in vivo DMPK properties as well as the in vivo antihypertensive effect in rats are presented.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Drug Discovery , Tetrahydronaphthalenes/chemistry , Tetrahydronaphthalenes/pharmacology , Animals , Calcium Channel Blockers/chemical synthesis , Calcium Channels/metabolism , Dose-Response Relationship, Drug , Molecular Conformation , Rats , Rats, Inbred SHR , Structure-Activity Relationship , Tetrahydronaphthalenes/chemical synthesis
16.
J Pharmacol Exp Ther ; 350(1): 130-43, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24769543

ABSTRACT

Treatment of pulmonary arterial hypertension with the endothelin receptor antagonist bosentan has been associated with transient increases in liver transaminases. Mechanistically, bosentan inhibits the bile salt export pump (BSEP) leading to an intrahepatic accumulation of cytotoxic bile salts, which eventually results in hepatocellular damage. BSEP inhibition by bosentan is amplified by its accumulation in the liver as bosentan is a substrate of organic anion-transporting polypeptide (OATP) transport proteins. The novel endothelin receptor antagonist macitentan shows a superior liver safety profile. Introduction of the less acidic sulfamide moiety and increased lipophilicity yield a hepatic disposition profile different from other endothelin receptor antagonists. Passive diffusion rather than OATP-mediated uptake is the driving force for macitentan uptake into the liver. Interaction with the sodium taurocholate cotransporting polypeptide and BSEP transport proteins involved in hepatic bile salt homeostasis is therefore limited due to the low intrahepatic drug concentrations. Evidence for this conclusion is provided by in vitro experiments in drug transporter-expressing cell lines, acute and long-term studies in rats and dogs, absence of plasma bile salt changes in healthy human volunteers after multiple dosing, and finally the liver safety profile of macitentan in the completed phase III morbidity/mortality SERAPHIN (Study with an Endothelin Receptor Antagonist in Pulmonary Arterial Hypertension to Improve Clinical Outcome) trial.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Bile Acids and Salts/blood , Liver/metabolism , Organic Anion Transporters, Sodium-Dependent/drug effects , Pyrimidines/pharmacokinetics , Pyrimidines/toxicity , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity , Symporters/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Animals , Bosentan , Cell Line , Cricetinae , Dogs , Dose-Response Relationship, Drug , Endothelin Receptor Antagonists , Hepatocytes , Humans , Male , Organic Anion Transporters/drug effects , Pyrimidines/adverse effects , Rats , Sulfonamides/adverse effects
17.
PLoS One ; 8(9): e74285, 2013.
Article in English | MEDLINE | ID: mdl-24069292

ABSTRACT

Ponesimod, a selective S1P1 receptor modulator, reduces the blood lymphocyte count in all tested species by preventing egress of T and B cells from thymus and peripheral lymphoid organs. In addition, ponesimod transiently affects heart rate and atrioventricular (AV) conduction in humans, effects not observed in mice, rats, and dogs with selective S1P1 receptor modulators, suggesting that the regulation of heart rate and rhythm is species dependent. In the present study, we used conscious guinea pigs implanted with a telemetry device to investigate the effects of single and multiple oral doses of ponesimod on ECG variables, heart rate, and blood pressure. Oral administration of ponesimod did not affect the sinus rate (P rate) but dose-dependently induced AV block type I to III. A single oral dose of 0.1 mg/kg had no effect on ECG variables, while a dose of 3 mg/kg induced AV block type III in all treated guinea pigs. Repeated oral dosing of 1 or 3 mg/kg ponesimod resulted in rapid desensitization, so that the second dose had no or a clearly reduced effect on ECG variables as compared with the first dose. Resensitization of the S1P1 receptor in the heart was concentration dependent. After desensitization had been induced by the first dose of ponesimod, the cardiac system remained desensitized as long as the plasma concentration was ≥75 ng/ml. By using a progressive up-titration regimen, the first-dose effect of ponesimod on heart rate and AV conduction was significantly reduced due to desensitization of the S1P1 receptor. In summary, conscious guinea pigs implanted with a telemetry device represent a useful model to study first-dose effects of S1P1 receptor modulators on heart rate and rhythm. This knowledge was translated to a dosing regimen of ponesimod to be tested in humans to avoid or significantly reduce the first-dose effects.


Subject(s)
Drug Tolerance , Heart/drug effects , Thiazoles/pharmacology , Administration, Oral , Animals , Atrioventricular Block/chemically induced , Atrioventricular Node/drug effects , Blood Pressure/drug effects , Electrocardiography , Guinea Pigs , Heart Rate/drug effects , Male , Receptors, Lysosphingolipid/metabolism , Thiazoles/administration & dosage , Thiazoles/adverse effects
18.
J Infect Dis ; 206(5): 735-43, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22732921

ABSTRACT

The increasing spread of drug-resistant malaria strains underscores the need for new antimalarial agents with novel modes of action (MOAs). Here, we describe a compound representative of a new class of antimalarials. This molecule, ACT-213615, potently inhibits in vitro erythrocytic growth of all tested Plasmodium falciparum strains, irrespective of their drug resistance properties, with half-maximal inhibitory concentration (IC(50)) values in the low single-digit nanomolar range. Like the clinically used artemisinins, the compound equally and very rapidly affects all 3 asexual erythrocytic parasite stages. In contrast, microarray studies suggest that the MOA of ACT-213615 is different from that of the artemisinins and other known antimalarials. ACT-213615 is orally bioavailable in mice, exhibits activity in the murine Plasmodium berghei model and efficacy comparable to that of the reference drug chloroquine in the recently established P. falciparum SCID mouse model. ACT-213615 represents a new class of potent antimalarials that merits further investigation for its clinical potential.


Subject(s)
Antimalarials/pharmacology , Malaria/drug therapy , Parasitemia/drug therapy , Piperazines/pharmacology , Plasmodium berghei/drug effects , Animals , Antimalarials/pharmacokinetics , Biological Availability , Cell Line , Erythrocytes/parasitology , Inhibitory Concentration 50 , Malaria/blood , Malaria/parasitology , Mice , Parasitemia/parasitology , Piperazines/pharmacokinetics , Rats
20.
J Med Chem ; 52(12): 3689-702, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19358611

ABSTRACT

Starting from known piperidine renin inhibitors, a new series of 3,9-diazabicyclo[3.3.1]nonene derivatives was rationally designed and prepared. Optimization of the positions 3, 6, and 7 of the diazabicyclonene template led to potent renin inhibitors. The substituents attached at the positions 6 and 7 were essential for the binding affinity of these compounds for renin. The introduction of a substituent attached at the position 3 did not modify the binding affinity but allowed the modulation of the ADME properties. Our efforts led to the discovery of compound (+)-26g that inhibits renin with an IC(50) of 0.20 nM in buffer and 19 nM in plasma. The pharmacokinetics properties of this and other similar compounds are discussed. Compound (+)-26g is well absorbed in rats and efficacious at 10 mg/kg in vivo.


Subject(s)
Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/pharmacology , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Renin/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Conformation , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...