Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Cyst Fibros ; 19(2): 292-298, 2020 03.
Article in English | MEDLINE | ID: mdl-31594690

ABSTRACT

BACKGROUND: Triple combinations of cystic fibrosis (CF) transmembrane conductance regulator (CFTR) modulators demonstrate enhanced clinical efficacy in CF patients with F508del mutation, compared with modest effects of dual combinations. GLPG2737 was developed as a novel corrector for triple combination therapy. METHODS: This multicenter, randomized, double-blind, placebo-controlled, phase 2a study evaluated GLPG2737 in F508del homozygous subjects who had been receiving lumacaftor 400mg/ivacaftor 250mg for ≥12weeks. The primary outcome was change from baseline in sweat chloride concentration. Other outcomes included assessment of pulmonary function, respiratory symptoms, safety, tolerability, and pharmacokinetics. RESULTS: Between November 2017 and April 2018, 22 subjects were enrolled and randomized to oral GLPG2737 (75mg; n=14) or placebo (n=8) capsules twice daily for 28days. A significant decrease from baseline in mean sweat chloride concentration occurred at day 28 for GLPG2737 versus placebo (least-squares-mean difference-19.6mmol/L [95% confidence interval (CI) -36.0, -3.2], p=.0210). The absolute improvement, as assessed by least-squares-mean difference in change from baseline, in forced expiratory volume in 1s (percent predicted) at day 28 for GLPG2737 versus placebo was 3.4% (95% CI -0.5, 7.3). Respiratory symptoms in both groups remained stable. Mild/moderate adverse events occurred in 10 (71.4%) and 8 (100%) subjects receiving GLPG2737 and placebo, respectively. Lower exposures of GLPG2737 (and active metabolite M4) were observed than would be expected if administered alone (as lumacaftor induces CYP3A4). Lumacaftor and ivacaftor exposures were as expected. CONCLUSIONS: GLPG2737 was well tolerated and yielded significant decreases in sweat chloride concentration versus placebo in subjects homozygous for F508del receiving lumacaftor/ivacaftor, demonstrating evidence of increased CFTR activity when added to a potentiator-corrector combination. FUNDING: Galapagos NV. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov identifier, NCT03474042.


Subject(s)
Aminophenols , Aminopyridines , Benzodioxoles , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis , Quinolones , Respiratory Function Tests/methods , Adult , Aminophenols/administration & dosage , Aminophenols/adverse effects , Aminophenols/pharmacokinetics , Aminopyridines/administration & dosage , Aminopyridines/adverse effects , Aminopyridines/pharmacokinetics , Benzodioxoles/administration & dosage , Benzodioxoles/adverse effects , Benzodioxoles/pharmacokinetics , Chloride Channel Agonists/administration & dosage , Chloride Channel Agonists/adverse effects , Chloride Channel Agonists/pharmacokinetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/physiopathology , Drug Combinations , Female , Homozygote , Humans , Male , Mutation , Quinolones/administration & dosage , Quinolones/adverse effects , Quinolones/pharmacokinetics , Sweat/chemistry , Treatment Outcome
2.
J Cyst Fibros ; 18(5): 700-707, 2019 09.
Article in English | MEDLINE | ID: mdl-31056441

ABSTRACT

BACKGROUND: Several treatment approaches in cystic fibrosis (CF) aim to correct CF transmembrane conductance regulator (CFTR) function; the efficacy of each approach is dependent on the mutation(s) present. A need remains for more effective treatments to correct functional deficits caused by the F508del mutation. METHODS: Two placebo-controlled, phase 2a studies evaluated GLPG2222, given orally once daily for 29 days, in subjects homozygous for F508del (FLAMINGO) or heterozygous for F508del and a gating mutation, receiving ivacaftor (ALBATROSS). The primary objective of both studies was to assess safety and tolerability. Secondary objectives included assessment of pharmacokinetics, and of the effect of GLPG2222 on sweat chloride concentrations, pulmonary function and respiratory symptoms. RESULTS: Fifty-nine and 37 subjects were enrolled into FLAMINGO and ALBATROSS, respectively. Treatment-related treatment-emergent adverse events (TEAEs) were reported by 29.2% (14/48) of subjects in FLAMINGO and 40.0% (12/30) in ALBATROSS; most were mild to moderate in severity and comprised primarily respiratory, gastrointestinal, and infection events. There were no deaths or discontinuations due to TEAEs. Dose-dependent decreases in sweat chloride concentrations were seen in GLPG2222-treated subjects (maximum decrease in FLAMINGO: -17.6 mmol/L [GLPG2222 200 mg], p < 0.0001; ALBATROSS: -7.4 mmol/L [GLPG2222 300 mg], p < 0.05). No significant effects on pulmonary function or respiratory symptoms were reported. Plasma GLPG2222 concentrations in CF subjects were consistent with previous studies in healthy volunteers and CF subjects. CONCLUSIONS: GLPG2222 was well tolerated. Sweat chloride reductions support on-target enhancement of CFTR activity in subjects with F508del mutation(s). Significant improvements in clinical endpoints were not demonstrated. Observed safety results support further evaluation of GLPG2222, including in combination with other CFTR modulators. FUNDING: Galapagos NV. Clinical trial registration numbers FLAMINGO, NCT03119649; ALBATROSS, NCT03045523.


Subject(s)
Aminophenols , Benzoates , Benzopyrans , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis , Drug Therapy, Combination/methods , Quinolones , Respiratory Function Tests/methods , Sweat , Administration, Oral , Adult , Aminophenols/administration & dosage , Aminophenols/adverse effects , Benzoates/administration & dosage , Benzoates/adverse effects , Benzoates/pharmacokinetics , Benzopyrans/administration & dosage , Benzopyrans/adverse effects , Benzopyrans/pharmacokinetics , Biological Availability , Chloride Channel Agonists/administration & dosage , Chloride Channel Agonists/adverse effects , Chloride Channel Agonists/pharmacokinetics , Cystic Fibrosis/diagnosis , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Double-Blind Method , Drug Monitoring , Female , Humans , Male , Mutation , Quinolones/administration & dosage , Quinolones/adverse effects , Sweat/chemistry , Sweat/drug effects , Treatment Outcome
3.
J Nucl Med ; 60(8): 1140-1146, 2019 08.
Article in English | MEDLINE | ID: mdl-30877174

ABSTRACT

The 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) enzyme converts cortisone to cortisol and participates in the regulation of glucocorticoid levels in tissues. 11ß-HSD1 is expressed in the liver, kidney, adipose tissue, placenta, and brain. 11ß-HSD1 is a target for treatment of depression, anxiety, posttraumatic stress disorder, and also against age-related cognitive function and memory loss. In this study, we evaluated the radiotracer 11C-AS2471907 (3-(2-chlorophenyl)-4-(methyl-11C)-5-[2-[2,4,6-trifluorophenoxy]propan-2-yl]-4H-1,2,4-triazole) to image 11ß-HSD1 availability in the human brain with PET. Methods: Fifteen subjects were included in the study. All subjects underwent one 2-h scan after a bolus administration of 11C-AS2471907. Two subjects underwent an additional scan after blockade with the selective and high-affinity 11ß-HSD1 inhibitor ASP3662 to evaluate 11C-AS2471907 nondisplaceable distribution volume. Five subjects also underwent an additional scan to evaluate the within-day test-retest variability of 11C-AS2471907 volumes of distribution (VT). Results:11C-AS2471907 time-activity curves were best fitted by the 2-tissue-compartment (2TC) model. 11C-AS2471907 exhibited a regionally varying pattern of uptake throughout the brain. The VT of 11C-AS2471907 ranged from 3.7 ± 1.5 mL/cm3 in the caudate nucleus to 14.5 ± 5.3 mL/cm3 in the occipital cortex, with intermediate values in the amygdala, white matter, cingulum, insula, frontal cortex, putamen, temporal and parietal cortices, cerebellum, and thalamus (from lowest to highest VT). From the blocking scans, nondisplaceable distribution volume was determined to be 0.16 ± 0.04 mL/cm3 for 11C-AS2471907. Thus, nearly all uptake was specific and the binding potential ranged from 22 in the caudate to 90 in the occipital cortex. Test-retest variability of 2TC VT values was less than 10% in most large cortical regions (14% in parietal cortex) and ranged from 14% (cerebellum) to 51% (amygdala) in other regions. The intraclass correlation coefficient of 2TC VT values ranged from 0.55 in the white matter to 0.98 in the cerebellum. Conclusion:11C-AS2471907 has a high fraction of specific binding in vivo in humans and reasonable within-day reproducibility of binding parameters.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Brain/enzymology , Positron-Emission Tomography , Triazoles/pharmacology , Adult , Brain Mapping , Carbon Radioisotopes/analysis , Humans , Kinetics , Male , Middle Aged , Radiopharmaceuticals/analysis , Reference Standards , Reproducibility of Results , Tissue Distribution , Triazoles/analysis
4.
Clin Transl Sci ; 12(3): 291-301, 2019 05.
Article in English | MEDLINE | ID: mdl-30740895

ABSTRACT

Inhibition of the enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) represents a potential mechanism for improving pain conditions. ASP3662 is a potent and selective inhibitor of 11ß-HSD1. Two phase I clinical studies were conducted to assess the safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of single and multiple ascending doses of ASP3662 in healthy young and elderly non-Japanese and young Japanese subjects. Nonlinear, more than dose-proportional PKs were observed for ASP3662 after single-dose administration, particularly at lower doses (≤ 6 mg); the PKs at steady state were dose proportional, although the time to ASP3662 steady state was dose dependent at lower doses (≤ 2 mg). Similar PKs were observed among young Japanese, young non-Japanese, and elderly non-Japanese subjects. Specific inhibition of 11ß-HSD1 occurred after both single and multiple doses of ASP3662. A marked dissociation between PKs and PDs was observed after single but not multiple doses of ASP3662. ASP3662 was generally safe and well tolerated.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Benzamides/adverse effects , Benzamides/pharmacokinetics , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Triazoles/adverse effects , Triazoles/pharmacokinetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/urine , Adolescent , Adult , Aged , Benzamides/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Middle Aged , Triazoles/pharmacology , Young Adult
5.
Clin Pharmacol Drug Dev ; 7(8): 911-919, 2018 11.
Article in English | MEDLINE | ID: mdl-29683561

ABSTRACT

An open-label, 3-period study was conducted in 30 healthy postmenopausal women (mean age, 58.4 years) who received a single oral dose of atorvastatin 20 mg on day 1 (period 1), multiple daily dosing of bazedoxifene 40 mg on days 4-11 (period 2), and coadministration of atorvastatin 20 mg + bazedoxifene 40 mg on day 12 (period 3). Serial blood samples were collected (24 hours after bazedoxifene and 72 hours after atorvastatin) and assayed for bazedoxifene, atorvastatin, and its ortho-hydroxy and para-hydroxy metabolites. Pharmacokinetic parameters were calculated using noncompartmental methods. Bazedoxifene exposure was not altered with coadministration of atorvastatin 20 mg (Cmax and AUCss were within bioequivalence limits). Similarly, atorvastatin and ortho-hydroxyatorvastatin exposure was equivalent with or without coadministration with bazedoxifene. Para-hydroxyatorvastatin concentrations were below the limit of quantitation under both conditions. Cmax for atorvastatin and ortho-hydroxyatorvastatin was 14% and 18% lower, respectively, and Tmax was 20% and 34% longer, respectively, with the combination compared with atorvastatin alone. There were no serious adverse events, and no subjects discontinued the study because of safety. No clinically significant pharmacokinetic interaction was observed between bazedoxifene and atorvastatin or its active metabolites, indicating they may be safely coadministered without dosage adjustment.


Subject(s)
Atorvastatin/pharmacokinetics , Indoles/pharmacokinetics , Anticholesteremic Agents/blood , Anticholesteremic Agents/pharmacokinetics , Atorvastatin/blood , Cross-Over Studies , Drug Interactions , Female , Healthy Volunteers , Humans , Indoles/blood , Middle Aged , Postmenopause/blood , Selective Estrogen Receptor Modulators/blood , Selective Estrogen Receptor Modulators/pharmacokinetics
6.
Clin Pharmacol Drug Dev ; 7(4): 365-372, 2018 05.
Article in English | MEDLINE | ID: mdl-29443456

ABSTRACT

Bazedoxifene, a selective estrogen receptor modulator with proestrogenic effects on bone and lipid metabolism and antiestrogenic effects on the breast and endometrium, is a treatment option for osteoporosis in postmenopausal women. It is extensively metabolized by the liver; therefore, a decrease in liver function was expected to decrease bazedoxifene clearance. This single-dose, open-label, inpatient/outpatient, nonrandomized study assessed the pharmacokinetics of bazedoxifene 20 mg in 18 postmenopausal women with hepatic impairment and 18 matched healthy postmenopausal women. Bazedoxifene elimination was slower, and exposure was higher, in hepatically impaired subjects compared with healthy subjects. In subjects with severe (Child-Pugh C) liver impairment, bazedoxifene mean half-life was 50% longer than that of healthy subjects. Area under the concentration-time curve geometric mean ratios (90%CI) for Child-Pugh A, B, and C liver impairment vs healthy subjects were 243% (156-379), 209% (135-326), and 368% (236-572), respectively. Although there were no severe adverse events in this study, bazedoxifene use in patients with hepatic impairment is not recommended.


Subject(s)
Indoles/adverse effects , Indoles/pharmacokinetics , Liver Diseases/blood , Postmenopause/blood , Adult , Aged , Area Under Curve , Case-Control Studies , Female , Half-Life , Humans , Indoles/administration & dosage , Liver Diseases/complications , Middle Aged
7.
Clin Pharmacol Drug Dev ; 7(4): 358-364, 2018 05.
Article in English | MEDLINE | ID: mdl-29389076

ABSTRACT

The purpose of this article was to evaluate the potential for a pharmacokinetic interaction between bazedoxifene and ibuprofen. In a randomized crossover study, 12 healthy postmenopausal women (aged 45-65 years) received either a single oral dose of ibuprofen (600-mg tablet), bazedoxifene (20-mg capsule), or both ibuprofen and bazedoxifene during the 3 treatment periods. Serial blood samples were collected for pharmacokinetic analyses. There was no relationship between the UGT1A1 genotype and bazedoxifene clearance. The 90% log-transformed confidence intervals (CIs) for bazedoxifene Cmax , 96% to 144%, and AUC, 85% to 134%, were slightly above the bioequivalence limits of 80% to 125%. The 90% log-transformed CIs for ibuprofen pharmacokinetic parameters were within these limits (Cmax , 92%-122%; AUC, 94%-106%). The increase in bazedoxifene plasma concentrations when combined with ibuprofen versus bazedoxifene alone is unlikely to be clinically significant. The lack of interaction between bazedoxifene and ibuprofen suggests that they may be coadministered without dose adjustment.


Subject(s)
Glucuronosyltransferase/genetics , Ibuprofen/pharmacokinetics , Indoles/pharmacokinetics , Postmenopause/blood , Administration, Oral , Area Under Curve , Biological Availability , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Female , Healthy Volunteers , Humans , Ibuprofen/administration & dosage , Indoles/administration & dosage , Middle Aged
8.
Clin Ther ; 35(6): 782-94, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23623756

ABSTRACT

BACKGROUND: Many antidepressants are extensively metabolized in the liver, requiring dose adjustments in individuals with hepatic impairment. Clinical studies indicate that the serotonin-norepinephrine reuptake inhibitor desvenlafaxine is metabolized primarily via glucuronidation, and ∼45% is eliminated unchanged in urine. OBJECTIVE: The objectives of this study were to assess the pharmacokinetic profile, safety, and tolerability of desvenlafaxine in adults with chronic Child-Pugh class A, B, and C hepatic impairment. METHODS: Subjects (aged 18-65 years) with mild (Child-Pugh class A, n = 8), moderate (Child-Pugh class B, n = 8), and severe (Child-Pugh class C, n = 8) hepatic impairment and 12 healthy matched subjects received a single 100-mg oral dose of desvenlafaxine. Disposition of (R)-, (S)-, and (R+S)-enantiomers of desvenlafaxine were examined in plasma and urine. Geometric least squares (GLS) mean ratios and 90% CIs for AUC, AUC0-τ, Cmax, and Cl/F were calculated; comparisons were made by using a 1-factor ANOVA. Safety was evaluated according to adverse events, physical examination, vital signs, and laboratory assessments. RESULTS: Healthy participants had a mean age of 51 years (range, 36-62 years) and weight of 79.1 kg (range, 52.5-105.0 kg); hepatically impaired participants had a mean age of 52 years (range, 31-65 years) and weight of 80.9 kg (range, 50.2-119.5 kg). In both groups, 67% of participants were male. No statistically significant differences (≥50%) in the disposition of desvenlafaxine were detected between hepatically impaired patients and healthy subjects based on GLS mean ratios for Cmax, AUC0-τ, AUC, or Cl/F (P > 0.05 for each comparison). Median Tmax was similar for all groups (range, 6-9 hours). A nonsignificant increase was observed for desvenlafaxine exposure in patients with moderate or severe hepatic impairment (GLS mean ratios [90% CIs] for AUC, 31% [93.2-184], 35% [96.5-190], respectively). The most common adverse events were nausea (n = 2, healthy subjects; n = 3, hepatically impaired subjects) and vomiting (n = 1, healthy subjects; n = 2, hepatically impaired subjects). CONCLUSIONS: A single 100-mg dose of desvenlafaxine was well tolerated in healthy subjects and hepatically impaired patients. A mild increase in exposure was observed for moderate and severe hepatically impaired subjects (Child-Pugh class B and C).


Subject(s)
Antidepressive Agents/administration & dosage , Cyclohexanols/administration & dosage , Liver Diseases/metabolism , Liver/drug effects , Neurotransmitter Uptake Inhibitors/administration & dosage , Adolescent , Adult , Aged , Antidepressive Agents/adverse effects , Antidepressive Agents/pharmacokinetics , Cyclohexanols/adverse effects , Cyclohexanols/pharmacokinetics , Desvenlafaxine Succinate , Female , Humans , Liver/metabolism , Male , Middle Aged , Neurotransmitter Uptake Inhibitors/adverse effects , Neurotransmitter Uptake Inhibitors/pharmacokinetics , Young Adult
9.
J Clin Pharmacol ; 51(1): 93-101, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20308689

ABSTRACT

Tigecycline belongs to a new class of tetracyclines, the glycylcyclines, less than 20% of which is metabolized in the liver. Twenty-five patients with cirrhosis with varying degrees of functional hepatic reserve (Child-Pugh A, n = 10; B, n = 10; C, n = 5) and 23 healthy adults, matched by age, sex, weight, and smoking habits, received 100 mg of tigecycline infused intravenously over 60 minutes. Serum and urine samples were collected up to 120 hours after dosing. Pharmacokinetic data were derived using noncompartmental methods. The most common treatment-emergent adverse events in healthy volunteers were nausea (56.5%), vomiting (21.7%), and headache (21.7%) and in the patients with cirrhosis, albuminuria (12%). Mean (± 1 SD) tigecycline clearance values were 29.8 ± 11.3 L/h in healthy subjects and 31.2 ± 13.9 L/h (Child-Pugh A), 22.1 ± 9.3 L/h (Child-Pugh B), and 13.5 ± 2.7 L/h (Child-Pugh C) in the patients. A single intravenous dose of tigecycline 100 mg was safe and well-tolerated in patients with cirrhosis with varying degrees of hepatic functional reserve. No adjustment of tigecycline maintenance dosage is warranted in patients with compensated or moderately decompensated cirrhosis; doses should be reduced by 50%, to 25 mg, every 12 hours in patients with severely decompensated disease.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Liver Cirrhosis/complications , Minocycline/analogs & derivatives , Adult , Anti-Bacterial Agents/adverse effects , Case-Control Studies , Female , Humans , Infusions, Intravenous , Liver Cirrhosis/pathology , Male , Middle Aged , Minocycline/adverse effects , Minocycline/pharmacokinetics , Severity of Illness Index , Tigecycline
SELECTION OF CITATIONS
SEARCH DETAIL
...