Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 33
Filtrer
1.
J Med Chem ; 67(18): 16692-16711, 2024 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-39276085

RÉSUMÉ

IL-17, a pro-inflammatory cytokine produced mainly by Th17 cells, is involved in the immune response to fungal and bacterial infections, whereas its aberrant production is associated with autoimmune and inflammatory diseases. IL-17 blocking antibodies like secukinumab (Cosentyx) have been developed and are used to treat conditions like psoriasis, psoriatic arthritis, and ankylosing spondylitis. Recently, the low molecular weight IL-17 inhibitor LY3509754 entered the clinic but was discontinued in Phase 1 due to adverse effects. In this study, we explored the replacements of furazan moiety posing a potential toxicology risk in LY3509754. By exploring replacements such as heterocycles as amide-isosteres as well as α-F-acrylamides, two compounds (18 and 26) were identified. Both compounds effectively reduced knee swelling in a rat arthritis model. However, early rat and dog toxicity studies revealed adverse findings, preventing their further development and indicating that furazan might not be responsible for the adverse effects of LY3509754.


Sujet(s)
Arthrite expérimentale , Interleukine-17 , Oxadiazoles , Animaux , Interleukine-17/antagonistes et inhibiteurs , Interleukine-17/métabolisme , Oxadiazoles/composition chimique , Oxadiazoles/pharmacologie , Oxadiazoles/usage thérapeutique , Rats , Arthrite expérimentale/traitement médicamenteux , Chiens , Découverte de médicament , Mâle , Relation structure-activité , Acrylates/composition chimique , Acrylates/pharmacologie , Acrylates/usage thérapeutique , Femelle , Humains
2.
J Med Chem ; 67(15): 12807-12818, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39018425

RÉSUMÉ

30 covalent drugs were used to assess clearance (CL) prediction reliability in animals and humans. In animals, marked CL underprediction was observed using cryopreserved hepatocytes or liver microsomes (LMs) supplemented for cytochrome P450 activity. Improved quantitative performance was observed by combining metabolic stability data from LMs and liver S9 fractions, the latter supplemented with reduced glutathione for glutathione transferase activity. While human LMs provided reliable human CL predictions, prediction statistics were improved further by incorporating S9 stability data. CL predictions with allometric scaling were less robust compared to in vitro drug metabolism methods; the best results were obtained using the fu-corrected intercept model. Human volume of distribution (Vd) was well predicted using allometric scaling of animal pharmacokinetic data; the most reliable results were achieved using simple allometric scaling of unbound Vd values. These results provide a quantitative framework to guide appropriate method selection for human PK prediction with covalent drugs.


Sujet(s)
Hépatocytes , Microsomes du foie , Humains , Animaux , Microsomes du foie/métabolisme , Hépatocytes/métabolisme , Préparations pharmaceutiques/métabolisme , Préparations pharmaceutiques/composition chimique , Cytochrome P-450 enzyme system/métabolisme , Administration par voie intraveineuse , Pharmacocinétique
3.
Hepatology ; 78(4): 1223-1239, 2023 10 01.
Article de Anglais | MEDLINE | ID: mdl-37162151

RÉSUMÉ

BACKGROUND AND AIMS: With distinct mechanisms of action, the combination of tropifexor (TXR) and cenicriviroc (CVC) may provide an effective treatment for NASH. This randomized, multicenter, double-blind, phase 2b study assessed the safety and efficacy of TXR and CVC combination, compared with respective monotherapies. APPROACH AND RESULTS: Patients (N = 193) were randomized 1:1:1:1 to once-daily TXR 140 µg (TXR 140 ), CVC 150 mg (CVC), TXR 140 µg + CVC 150 mg (TXR 140 + CVC), or TXR 90 µg + CVC 150 mg (TXR 90 + CVC) for 48 weeks. The primary and secondary end points were safety and histological improvement, respectively. Rates of adverse events (AEs) were similar across treatment groups. Pruritus was the most frequently experienced AE, with highest incidence in the TXR 140 group (40.0%). In TXR and combination groups, alanine aminotransferase (ALT) decreased from baseline to 48 weeks (geometric mean change: -21%, TXR 140 ; -16%, TXR 140 + CVC; -13%, TXR 90 + CVC; and +17%, CVC). Reductions in body weight observed at week 24 (mean changes from baseline: TXR 140 , -2.5 kg; TXR 140 + CVC, -1.7 kg; TXR 90 + CVC, -1.0 kg; and CVC, -0.1 kg) were sustained to week 48. At least 1-point improvement in fibrosis stage/steatohepatitis resolution without worsening of fibrosis was observed in 32.3%/25.8%, 31.6%/15.8%, 29.7%/13.5%, and 32.5%/22.5% of patients in the TXR 140 , CVC, TXR 140 + CVC, and TXR 90 + CVC groups, respectively. CONCLUSIONS: The safety profile of TXR + CVC combination was similar to respective monotherapies, with no new signals. TXR monotherapy showed sustained ALT and body weight decreases. No substantial incremental efficacy was observed with TXR + CVC combination on ALT, body weight, or in histological end points compared with monotherapy.


Sujet(s)
Stéatose hépatique non alcoolique , Humains , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/complications , Méthode en double aveugle , Résultat thérapeutique , Fibrose , Poids
4.
Clin Pharmacol Drug Dev ; 12(2): 122-131, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36495282

RÉSUMÉ

This open-label, randomized, 3-treatment, 3-period, 6-sequence, crossover study in healthy subjects compared the pharmacokinetic and pharmacodynamic properties of a lipid-based (soft gelatin capsule) prototype final market image (pFMI) formulation of tropifexor (90-µg) to its clinical service form (CSF) and assessed the food effect for the pFMI formulation. In the fasted state, drug exposure was higher for the pFMI. The geometric mean ratios for pFMI versus CSF of peak concentration and area under the concentration-time curve were 2.0 and 1.5, respectively. No food effect was apparent for the pFMI formulation, and the geometric mean ratios for pFMI fed versus pFMI fasted of peak concentration and area under concentration-time curve were 1.0 and 1.0 respectively. Despite having lower systemic exposure, the CSF formulation provided a higher pharmacological response for the gut biomarker fibroblast growth factor 19. Under fasted conditions, fibroblast growth factor 19 maximum change from baseline serum concentration after drug administration and area under the change from baseline serum concentration-time curve from time 0 to 24 hours were 36% for CSF and 12% for FMI. For a second biomarker, serum 7-alpha hydroxy-4-cholest-3-one, the pharmacological activity was comparable between CSF (fasted) and pFMI (both fasted and fed states). The pFMI offers advantages over the CSF in terms of insensitivity to food effect, lower intersubject variability, and overcoming solubility limitations.


Sujet(s)
Interactions aliments-médicaments , Humains , Aire sous la courbe , Biodisponibilité , Études croisées , Volontaires sains
5.
J Med Chem ; 65(24): 16173-16203, 2022 12 22.
Article de Anglais | MEDLINE | ID: mdl-36399068

RÉSUMÉ

Rapid emergence of tumor resistance via RAS pathway reactivation has been reported from clinical studies of covalent KRASG12C inhibitors. Thus, inhibitors with broad potential for combination treatment and distinct binding modes to overcome resistance mutations may prove beneficial. JDQ443 is an investigational covalent KRASG12C inhibitor derived from structure-based drug design followed by extensive optimization of two dissimilar prototypes. JDQ443 is a stable atropisomer containing a unique 5-methylpyrazole core and a spiro-azetidine linker designed to position the electrophilic acrylamide for optimal engagement with KRASG12C C12. A substituted indazole at pyrazole position 3 results in novel interactions with the binding pocket that do not involve residue H95. JDQ443 showed PK/PD activity in vivo and dose-dependent antitumor activity in mouse xenograft models. JDQ443 is now in clinical development, with encouraging early phase data reported from an ongoing Phase Ib/II clinical trial (NCT04699188).


Sujet(s)
Tumeurs , Protéines proto-oncogènes p21(ras) , Animaux , Humains , Souris , Modèles animaux de maladie humaine , Conception de médicament , Mutation , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Pyrazoles/pharmacologie , Pyrazoles/usage thérapeutique
6.
Clin Pharmacol Drug Dev ; 11(11): 1253-1263, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35962468

RÉSUMÉ

Tropifexor, a farnesoid X receptor agonist, is currently under clinical development for the treatment of nonalcoholic steatohepatitis. Tropifexor undergoes glucuronidation by uridine 5'-diphosphoglucuronosyltransferase (UGT) 1A1 and oxidation by cytochrome P450 (CYP) 3A4, as reported in in vitro studies. Here, we report the results from 2 drug-drug interaction studies. Study 1 enrolled 20 healthy subjects to investigate the effect of the UGT1A1 inhibitor atazanavir (ATZ) on tropifexor pharmacokinetics (PK). Study 2 had 2 cohorts with 16 healthy subjects each to investigate the effect of the strong CYP3A4 inhibitor itraconazole and strong CYP3A4 inducer rifampin on the PK of tropifexor. Coadministration of ATZ reduced the maximum plasma concentration (Cmax ) of tropifexor by 40%; however, it did not lead to increased exposure of tropifexor (both area under the plasma concentration-time curve [AUC] from time 0 to the last quantifiable concentration [AUClast ] and AUC from time 0 to infinity [AUCinf ] reduced by only 10%), suggesting minor relevance of the UGT1A1 pathway for clearance of tropifexor and no expected drug-drug interactions based on UGT1A1 inhibition. Inhibition of CYP3A4 by itraconazole increased the Cmax of tropifexor by only 9% and exposure (both AUClast and AUCinf ) by 47%, suggesting a weak effect of strong CYP3A4 inhibitors on tropifexor PK. Inducing CYP3A4 with rifampin decreased Cmax (55%) and AUC (AUClast by 79% and AUCinf by 77%). Coadministration of tropifexor with either ATZ, itraconazole, or rifampin was well tolerated.


Sujet(s)
Cytochrome P-450 CYP3A , Itraconazole , Humains , Cytochrome P-450 CYP3A/métabolisme , Inhibiteurs du cytochrome P-450 CYP3A/pharmacologie , Interactions médicamenteuses , Volontaires sains , Rifampicine
7.
Cancer Discov ; 12(6): 1500-1517, 2022 06 02.
Article de Anglais | MEDLINE | ID: mdl-35404998

RÉSUMÉ

Covalent inhibitors of KRASG12C have shown antitumor activity against advanced/metastatic KRASG12C-mutated cancers, though resistance emerges and additional strategies are needed to improve outcomes. JDQ443 is a structurally unique covalent inhibitor of GDP-bound KRASG12C that forms novel interactions with the switch II pocket. JDQ443 potently inhibits KRASG12C-driven cellular signaling and demonstrates selective antiproliferative activity in KRASG12C-mutated cell lines, including those with G12C/H95 double mutations. In vivo, JDQ443 induces AUC exposure-driven antitumor efficacy in KRASG12C-mutated cell-derived (CDX) and patient-derived (PDX) tumor xenografts. In PDX models, single-agent JDQ443 activity is enhanced by combination with inhibitors of SHP2, MEK, or CDK4/6. Notably, the benefit of JDQ443 plus the SHP2 inhibitor TNO155 is maintained at reduced doses of either agent in CDX models, consistent with mechanistic synergy. JDQ443 is in clinical development as monotherapy and in combination with TNO155, with both strategies showing antitumor activity in patients with KRASG12C-mutated tumors. SIGNIFICANCE: JDQ443 is a structurally novel covalent KRASG12C inhibitor with a unique binding mode that demonstrates potent and selective antitumor activity in cell lines and in vivo models. In preclinical models and patients with KRASG12C-mutated malignancies, JDQ443 shows potent antitumor activity as monotherapy and in combination with the SHP2 inhibitor TNO155. This article is highlighted in the In This Issue feature, p. 1397.


Sujet(s)
Antienzymes , Indazoles , Tumeurs , Protéines proto-oncogènes p21(ras) , Antienzymes/composition chimique , Antienzymes/pharmacologie , Humains , Indazoles/composition chimique , Indazoles/pharmacologie , Mutation , Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Tumeurs/génétique , Protéines proto-oncogènes p21(ras)/antagonistes et inhibiteurs , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme
8.
J Clin Pharmacol ; 62(4): 520-531, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-34738233

RÉSUMÉ

Tropifexor, a non-bile acid farnesoid X receptor (FXR) agonist, has dose-proportional pharmacokinetics and no obvious major enterohepatic circulation. This open-label study investigated the effect of hepatic impairment (HI), as determined by Child-Pugh grade, on tropifexor's pharmacokinetics, safety, and tolerability following a 200-µg dose in the fasted state. Blood samples were collected through 168 hours after dosing for quantification and plasma protein-binding determination. Total tropifexor exposure was comparable across participants with HI vs those with normal hepatic function. Tropifexor was highly protein bound (>99%) in human plasma across participants of all groups. The average unbound fractions (percentage free) were 0.14% in participants with normal hepatic function and mild HI, which increased to 0.17% and 0.24% in participants with moderate and severe HI, respectively. Similar unbound drug exposure was noted in participants with mild HI and normal hepatic function. Participants with moderate HI (N = 8) had a 1.6-fold increase in unbound exposure (area under the plasma concentration-time curve from time 0 to infinity [AUCinf,u ]) and a 1.3-fold increase in maximal exposure (Cmax,u ) vs those with normal hepatic function (geometric mean ratio: AUCinf,u , 1.64 [90%CI, 1.25-2.16]; Cmax,u , 1.30 [90%CI, 0.96-1.76]). Participants with severe HI (N = 8) had a 1.6-fold increase in AUCinf,u (1.61 [90%CI, 1.04-2.49]) and comparable Cmax,u (1.02 [90%CI, 0.60-1.72]) compared to participants with normal hepatic function. Tropifexor was well tolerated. The relative insensitivity of tropifexor to HI offers the potential to treat patients with severe liver disease without dose adjustment.


Sujet(s)
Isoxazoles , Maladies du foie , Aire sous la courbe , Benzothiazoles , Humains , Maladies du foie/métabolisme
9.
J Med Chem ; 63(17): 9856-9875, 2020 09 10.
Article de Anglais | MEDLINE | ID: mdl-32856916

RÉSUMÉ

G-protein-coupled receptor SUCNR1 (succinate receptor 1 or GPR91) senses the citric cycle intermediate succinate and is implicated in various pathological conditions such as rheumatoid arthritis, liver fibrosis, or obesity. Here, we describe a novel SUCNR1 antagonist scaffold discovered by high-throughput screening. The poor permeation and absorption properties of the most potent compounds, which were zwitterionic in nature, could be improved by the formation of an internal salt bridge, which helped in shielding the two opposite charges and thus also the high polarity of zwitterions with separated charges. The designed compounds containing such a salt bridge reached high oral bioavailability and oral exposure. We believe that this principle could find a broad interest in the medicinal chemistry field as it can be useful not only for the modulation of properties in zwitterionic compounds but also in acidic or basic compounds with poor permeation.


Sujet(s)
Benzamides/pharmacologie , Phénylacétates/pharmacologie , Récepteurs couplés aux protéines G/antagonistes et inhibiteurs , Animaux , Benzamides/synthèse chimique , Benzamides/métabolisme , Benzamides/pharmacocinétique , Lignée cellulaire , Découverte de médicament , Humains , Mâle , Souris de lignée C57BL , Phénylacétates/synthèse chimique , Phénylacétates/métabolisme , Phénylacétates/pharmacocinétique , Liaison aux protéines , Rats , Récepteurs couplés aux protéines G/métabolisme , Électricité statique
10.
J Med Chem ; 62(23): 10816-10832, 2019 12 12.
Article de Anglais | MEDLINE | ID: mdl-31729873

RÉSUMÉ

Retinoic acid receptor-related orphan receptor gamma-t (RORγt) is considered to be the master transcription factor for the development of Th17 cells that produce proinflammatory cytokines such as IL-17A. Overproportionate Th17 cell abundance is associated with the pathogenesis of many inflammatory conditions including psoriasis. In a high-throughput fluorescence resonance energy transfer (FRET) screen, we identified compound 1 as a hit with promising lipophilic efficiency (LipE). Using structure-based drug design based on a number of X-ray cocrystal structures, we morphed this hit class into potent imidazoles, exemplified by compound 3. To improve the poor absorption, distribution, metabolism, and excretion (ADME) properties of neutral imidazoles, we extended our ligands with carboxylic acid substituents toward a polar, water-rich area of the protein. This highly lipophilicity-efficient modification ultimately led to the discovery of compound 14, a potent and selective inhibitor of RORγt with good ADME properties and excellent in vivo pharmacokinetics. This compound showed good efficacy in an in vivo delayed-type hypersensitivity pharmacology model in rats.


Sujet(s)
Hypersensibilité retardée/traitement médicamenteux , Imidazoles/pharmacologie , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/antagonistes et inhibiteurs , Administration par voie orale , Animaux , Relation dose-effet des médicaments , Conception de médicament , Femelle , Transfert d'énergie par résonance de fluorescence , Période , Imidazoles/composition chimique , Imidazoles/pharmacocinétique , Mâle , Modèles moléculaires , Structure moléculaire , Rats
11.
Xenobiotica ; 49(1): 13-21, 2019 Jan.
Article de Anglais | MEDLINE | ID: mdl-29299977

RÉSUMÉ

1. The utility of 1-aminobenzotriazole (ABT), incorporated in food, has been investigated as an approach for longer term inhibition of cytochrome P450 (P450) enzymes in mice. 2. In rats, ABT inhibits gastric emptying, to investigate this potential limitation in mice we examined the effect of ABT administration on the oral absorption of NVS-CRF38. Two hour prior oral treatment with 100 mg/kg ABT inhibited the oral absorption of NVS-CRF38, Tmax was 4 hours for ABT-treated mice compared to 0.5 hours in the control group. 3. A marked inhibition of hepatic P450 activity was observed in mice fed with ABT containing food pellets for 1 month. P450 activity, as measured by the oral clearance of antipyrine, was inhibited on day 3 (88% of control), week 2 (83% of control) and week 4 (80% of control). 4. Tmax values for antipyrine were comparable between ABT-treated mice and the control group, alleviating concerns about impaired gastric function. 5. Inclusion of ABT in food provides a minimally invasive and convenient approach to achieve longer term inhibition of P450 activity in mice. This model has the potential to enable pharmacological proof-of-concept studies for research compounds which are extensively metabolised by P450 enzymes.


Sujet(s)
Inhibiteurs des enzymes du cytochrome P-450/pharmacologie , Cytochrome P-450 enzyme system/métabolisme , Triazoles/pharmacologie , Administration par voie orale , Animaux , Souris , Oxazoles/métabolisme , Pyrazoles/métabolisme
12.
J Med Chem ; 61(15): 6724-6735, 2018 08 09.
Article de Anglais | MEDLINE | ID: mdl-29990434

RÉSUMÉ

The transcription factor RORγt is an attractive drug-target due to its role in the differentiation of IL-17 producing Th17 cells that play a critical role in the etiopathology of several autoimmune diseases. Identification of starting points for RORγt inverse agonists with good properties has been a challenge. We report the identification of a fragment hit and its conversion into a potent inverse agonist through fragment optimization, growing and merging efforts. Further analysis of the binding mode revealed that inverse agonism was achieved by an unusual mechanism. In contrast to other reported inverse agonists, there is no direct interaction or displacement of helix 12 observed in the crystal structure. Nevertheless, compound 9 proved to be efficacious in a delayed-type hypersensitivity (DTH) inflammation model in rats.


Sujet(s)
Découverte de médicament , Agonisme inverse des médicaments , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/agonistes , Animaux , Domaine catalytique , Modèles animaux de maladie humaine , Femelle , Inflammation/métabolisme , Modèles moléculaires , Rats
14.
ACS Med Chem Lett ; 8(5): 582-586, 2017 May 11.
Article de Anglais | MEDLINE | ID: mdl-28523115

RÉSUMÉ

Further optimization of an initial DP2 receptor antagonist clinical candidate NVP-QAV680 led to the discovery of a follow-up molecule 2-(2-methyl-1-(4-(methylsulfonyl)-2-(trifluoromethyl)benzyl)-1H-pyrrolo[2,3-b]pyridin-3-yl)acetic acid (compound 11, NVP-QAW039, fevipiprant), which exhibits improved potency on human eosinophils and Th2 cells, together with a longer receptor residence time, and is currently in clinical trials for severe asthma.

15.
JCI Insight ; 2(5): e91127, 2017 03 09.
Article de Anglais | MEDLINE | ID: mdl-28289717

RÉSUMÉ

Retinoic-acid-orphan-receptor-C (RORC) is a master regulator of Th17 cells, which are pathogenic in several autoimmune diseases. Genetic Rorc deficiency in mice, while preventing autoimmunity, causes early lethality due to metastatic thymic T cell lymphomas. We sought to determine whether pharmacological RORC inhibition could be an effective and safe therapy for autoimmune diseases by evaluating its effects on Th17 cell functions and intrathymic T cell development. RORC inhibitors effectively inhibited Th17 differentiation and IL-17A production, and delayed-type hypersensitivity reactions. In vitro, RORC inhibitors induced apoptosis, as well as Bcl2l1 and BCL2L1 mRNA downregulation, in mouse and nonhuman primate thymocytes, respectively. Chronic, 13-week RORC inhibitor treatment in rats caused progressive thymic alterations in all analyzed rats similar to those in Rorc-deficient mice prior to T cell lymphoma development. One rat developed thymic cortical hyperplasia with preneoplastic features, including increased mitosis and reduced IKAROS expression, albeit without skewed T cell clonality. In summary, pharmacological inhibition of RORC not only blocks Th17 cell development and related cytokine production, but also recapitulates thymic aberrations seen in Rorc-deficient mice. While RORC inhibition may offer an effective therapeutic principle for Th17-mediated diseases, T cell lymphoma with chronic therapy remains an apparent risk.


Sujet(s)
Récepteurs à l'acide rétinoïque/antagonistes et inhibiteurs , Cellules Th17/cytologie , Thymus (glande)/anatomopathologie , Animaux , Régulation négative , Femelle , Expression des gènes , Humains , Cellules Jurkat , Mâle , Souris , Souris de lignée C57BL , Rats , Rats de lignée LEW , Rat Sprague-Dawley , Récepteurs à l'acide rétinoïque/génétique , Cellules Th17/métabolisme
16.
ChemMedChem ; 11(24): 2640-2648, 2016 12 16.
Article de Anglais | MEDLINE | ID: mdl-27902884

RÉSUMÉ

Retinoic-acid-related orphan receptor γt (RORγt) is a key transcription factor implicated in the production of pro-inflammatory Th17 cytokines, which drive a number of autoimmune diseases. Despite diverse chemical series having been reported, combining high potency with a good physicochemical profile has been a very challenging task in the RORγt inhibitor field. Based on available chemical structures and incorporating in-house knowledge, a new series of triazolo- and imidazopyridine RORγt inverse agonists was designed. In addition, replacement of the terminal cyclopentylamide metabolic soft spot by five-membered heterocycles was investigated. From our efforts, we identified an optimal 6,7,8-substituted imidazo[1,2-a]pyridine core system and a 5-tert-butyl-1,2,4-oxadiazole as cyclopentylamide replacement leading to compounds 10 ((S)-N-(8-((4-(cyclopentanecarbonyl)-3-methylpiperazin-1-yl)methyl)-7-methylimidazo[1,2-a]pyridin-6-yl)-2-methylpyrimidine-5-carboxamide) and 33 ((S)-N-(8-((4-(5-(tert-butyl)-1,2,4-oxadiazol-3-yl)-3-methylpiperazin-1-yl)methyl)-7-methylimidazo[1,2-a]pyridin-6-yl)-2-methylpyrimidine-5-carboxamide). Both derivatives showed good pharmacological potencies in biochemical and cell-based assays combined with excellent physicochemical properties, including low to medium plasma protein binding across species. Finally, 10 and 33 were shown to be active in a rodent pharmacokinetic/pharmacodynamic (PK/PD) model after oral gavage at 15 mg kg-1 , lowering IL-17 cytokine production in ex vivo antigen recall assays.


Sujet(s)
Agonisme inverse des médicaments , Imidazoles , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/agonistes , Pyridines/synthèse chimique , Récepteurs à l'acide rétinoïque/agonistes , Triazoles , Animaux , Sites de fixation , Cellules cultivées , Cristallographie aux rayons X , Humains , Imidazoles/synthèse chimique , Imidazoles/composition chimique , Imidazoles/pharmacologie , Concentration inhibitrice 50 , Interleukine-17/sang , Structure moléculaire , Liaison aux protéines/effets des médicaments et des substances chimiques , Pyridines/composition chimique , Pyridines/pharmacologie , Rats , Triazoles/synthèse chimique , Triazoles/composition chimique , Triazoles/pharmacologie
17.
J Med Chem ; 59(17): 7901-14, 2016 09 08.
Article de Anglais | MEDLINE | ID: mdl-27502700

RÉSUMÉ

A series of potent PDGFR inhibitors has been identified. The series was optimized for duration of action in the lung. A novel kinase occupancy assay was used to directly measure target occupancy after i.t. dosing. Compound 25 shows 24 h occupancy of the PDGFR kinase domain, after a single i.t. dose and has efficacy at 0.03 mg/kg, in the rat moncrotaline model of pulmonary arterial hypertension. Examination of PK/PD data from the optimization effort has revealed in vitro:in vivo correlations which link duration of action in vivo with low permeability and high basicity and demonstrate that nonspecific binding to lung tissue increases with lipophilicity.


Sujet(s)
Remodelage des voies aériennes/effets des médicaments et des substances chimiques , Hypertension pulmonaire/traitement médicamenteux , Nicotinamide/analogues et dérivés , Pyrazoles/composition chimique , Récepteurs aux facteurs de croissance dérivés des plaquettes/antagonistes et inhibiteurs , Remodelage vasculaire/effets des médicaments et des substances chimiques , Administration par inhalation , Animaux , Lignée cellulaire , Prolifération cellulaire , Hypertension pulmonaire/anatomopathologie , Poumon/vascularisation , Membrane artificielle , Simulation de docking moléculaire , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/anatomopathologie , Nicotinamide/synthèse chimique , Nicotinamide/composition chimique , Nicotinamide/pharmacologie , Perméabilité , Protéines proto-oncogènes c-kit/antagonistes et inhibiteurs , Protéines proto-oncogènes c-kit/composition chimique , Pyrazoles/synthèse chimique , Pyrazoles/pharmacologie , Rats , Récepteur au PDGF alpha/antagonistes et inhibiteurs , Récepteur au PDGF alpha/composition chimique , Récepteur au PDGF bêta/antagonistes et inhibiteurs , Récepteur au PDGF bêta/composition chimique , Récepteurs aux facteurs de croissance dérivés des plaquettes/composition chimique , Relation structure-activité
18.
Drug Metab Dispos ; 44(8): 1213-6, 2016 08.
Article de Anglais | MEDLINE | ID: mdl-27271368

RÉSUMÉ

The effectiveness of controlled release 1-aminobenzotriazole (ABT) administration to inhibit cytochrome P450 (P450) enzymes has been evaluated in mice. To maximize the duration of P450 inhibition in vivo, ABT was administered via an osmotic pump. The degree of P450 inhibition was compared with that achieved with a single bolus dose of ABT. Two-hour prior subcutaneous treatment of mice with ABT (50 mg/kg) inhibited antipyrine clearance by 88%. A less pronounced inhibitory effect (29% reduction in clearance) was observed when ABT was administered 24-hours before antipyrine administration, indicating partial restoration of P450 activity during this longer pretreatment time. The duration of ABT in mice was very short (mean residence time = 1.7 hours) after subcutaneous bolus administration. When the inhibitor was delivered by an osmotic pump, maximum blood concentrations of the inhibitor were observed 24 hours after device implantation and were maintained at steady state for 6 days. Inhibition of P450 activity, as measured by antipyrine clearance, was confirmed at 24 hours and 120 hours after pump implantation, highlighting the utility of this method as a longer-term model for P450 inhibition in mice. The magnitude of P450 inhibition in ABT-treated mice was compared with that in hepatic P450 reductase null mice and both models were comparable. In vivo ABT administration by an osmotic pump offers an effective approach for longer-term P450 inhibition in mice and avoids the necessity for multiple dosing of the inhibitor.


Sujet(s)
Phénazone/pharmacocinétique , Inhibiteurs des enzymes du cytochrome P-450/administration et posologie , Cytochrome P-450 enzyme system/déficit , Pompes à perfusion implantables , Foie/effets des médicaments et des substances chimiques , Triazoles/administration et posologie , Animaux , Phénazone/administration et posologie , Phénazone/sang , Inhibiteurs des enzymes du cytochrome P-450/sang , Cytochrome P-450 enzyme system/génétique , Génotype , Perfusions sous-cutanées , Injections sous-cutanées , Foie/enzymologie , Mâle , Souris knockout , Pression osmotique , Phénotype , Triazoles/sang
19.
J Med Chem ; 57(12): 5129-40, 2014 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-24884675

RÉSUMÉ

A high throughput screening campaign identified 5-(2-chlorophenyl)indazole compound 4 as an antagonist of the transient receptor potential A1 (TRPA1) ion channel with IC50 = 1.23 µM. Hit to lead medicinal chemistry optimization established the SAR around the indazole ring system, demonstrating that a trifluoromethyl group at the 2-position of the phenyl ring in combination with various substituents at the 6-position of the indazole ring greatly contributed to improvements in vitro activity. Further lead optimization resulted in the identification of compound 31, a potent and selective antagonist of TRPA1 in vitro (IC50 = 0.015 µM), which has moderate oral bioavailability in rodents and demonstrates robust activity in vivo in several rodent models of inflammatory pain.


Sujet(s)
Indazoles/composition chimique , Protéines de tissu nerveux/antagonistes et inhibiteurs , Canaux cationiques TRP/antagonistes et inhibiteurs , Administration par voie orale , Analgésiques/composition chimique , Analgésiques/pharmacocinétique , Analgésiques/pharmacologie , Animaux , Anti-inflammatoires/composition chimique , Anti-inflammatoires/pharmacocinétique , Anti-inflammatoires/pharmacologie , Biodisponibilité , Cellules CHO , Canaux calciques , Cricetulus , Adjuvant Freund , Humains , Hyperalgésie/induit chimiquement , Hyperalgésie/traitement médicamenteux , Indazoles/pharmacocinétique , Indazoles/pharmacologie , Mâle , Souris de lignée C57BL , Moutarde (plante) , Huiles végétales , Rat Wistar , Spécificité d'espèce , Relation structure-activité , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire , Canaux cationiques TRPC/antagonistes et inhibiteurs
20.
Drug Metab Dispos ; 42(7): 1117-24, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-24727486

RÉSUMÉ

The simultaneous effects of the cytochrome P450 inhibitor 1-aminobenzotriazole (ABT) on inhibition of in vivo metabolism and gastric emptying were evaluated with the test compound 7-(3,5-dimethyl-1H-1,2,4-triazol-1-yl)-3-(4-methoxy-2-methylphenyl)-2,6-dimethylpyrazolo[5,1-b]oxazole(NVS-CRF38), a novel corticotropin releasing factor receptor 1 (CRF1) antagonist with low water solubility, and the reference compound midazolam with high water solubility in rats. Pretreatment of rats with 100 mg/kg oral ABT administered 2 hours before a semisolid caloric test meal markedly delayed gastric emptying. ABT increased stomach weights by 2-fold; this is likely attributable to a prosecretory effect because stomach concentrations of bilirubin were comparable in ABT and control groups. ABT administration decreased the initial systemic exposure of orally administered NVS-CRF38 and increased Tmax 40-fold, suggesting gastric retention and delayed oral absorption. ABT increased the initial systemic exposure of midazolam, however for orally (but not subcutaneously) administered midazolam, extensive variability in plasma-concentration time profiles was apparent. Careful selection of administration routes is recommended for ABT use in vivo, variable oral absorption of coadministered compounds can be expected due to a disturbance of gastrointestinal transit.


Sujet(s)
Inhibiteurs des enzymes du cytochrome P-450/pharmacologie , Vidange gastrique/effets des médicaments et des substances chimiques , Triazoles/pharmacologie , Administration par voie orale , Animaux , Femelle , Rats , Rat Sprague-Dawley , Rat Wistar , Triazoles/administration et posologie , Triazoles/pharmacocinétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE