Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Pediatr Pulmonol ; 58(3): 908-917, 2023 03.
Article in English | MEDLINE | ID: mdl-36507572

ABSTRACT

BACKGROUND: The PATENT-CHILD study investigated riociguat in children aged ≥ 6 to <18 years with pulmonary arterial hypertension (PAH) treated with tablets or an oral pediatric suspension based on bodyweight-adjusted dosing of up to 2.5 mg three times daily. PATENT-CHILD demonstrated an acceptable riociguat safety profile and individual plasma concentrations in pediatric patients were consistent with those in adult patients. METHODS: Using the data set from PATENT-CHILD and building on existing population pharmacokinetic (PK) models for riociguat and its major metabolite (M1) in adults with PAH, a coupled riociguat-M1 PK model was developed. The final model developed incorporated a one-compartment model for riociguat, coupled to a one-compartment model for M1, allowing for presystemic formation of M1. It included allometric scaling exponents for bodyweight. RESULTS: Apparent clearance of riociguat was similar in children and adult patients with PAH (median [interquartile range] 2.20 [1.75-3.44] and 2.08 L/h [1.55-2.97]). Factors contributing to lower PK exposure were lower riociguat maintenance dose in PATENT-CHILD, and a higher riociguat clearance in some adolescent patients, compared with adult patients. No effects of formulation, sex, or age on riociguat PK were observed. An exploratory PK/pharmacodynamics analysis found the increase in 6-min walking distance in pediatric patients treated with riociguat was not related to riociguat PK. CONCLUSIONS: Body size is the main determinant of PK in growing children, and the model supports clinical data that, for children weighing < 50 kg, a bodyweight-adjusted dose of riociguat should be used to achieve a similar exposure to that observed in adults with PAH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Adult , Adolescent , Humans , Child , Pulmonary Arterial Hypertension/drug therapy , Hypertension, Pulmonary/drug therapy , Treatment Outcome , Familial Primary Pulmonary Hypertension/drug therapy , Drug Administration Schedule
2.
Basic Clin Pharmacol Toxicol ; 128(3): 511-524, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33232579

ABSTRACT

Molidustat is an oral inhibitor of hypoxia-inducible factor (HIF) prolyl-hydroxylase enhancing the erythropoietin (EPO) response to HIF; it is in clinical development for the treatment of anaemia related to chronic kidney disease. The predominant role of glucuronidation for molidustat clearance (formation of N-glucuronide metabolite M1) and subsequent renal excretion was confirmed in a human mass balance study, with about 85% of the drug being excreted as M1 in urine. The inhibitory effects of 176 drugs and xenobiotics from various compound classes on the UGT-mediated glucuronidation of molidustat in human liver microsomes (HLMs) were investigated. Based on preclinical findings, glucuronidation of molidustat was predominantly mediated by the 5'-diphospho-glucuronosyltransferase (UGT) isoform UGT1A1. Therefore, atazanavir, which is a potent inhibitor of UGT1A1, was chosen for the evaluation of pharmacokinetics and EPO release following a single oral dose of 25 mg molidustat. Molidustat exposure increased about twofold upon coadministration with atazanavir when considering area under plasma concentration-time curve from zero to infinity (AUC) and maximum plasma concentration (Cmax ). Baseline-corrected increase of EPO was 14% and 34% for Cmax and AUC (calculated over 24 hours), respectively. Coadministration of molidustat and atazanavir was well tolerated.


Subject(s)
Atazanavir Sulfate/pharmacology , Glucuronides/metabolism , Glucuronosyltransferase/physiology , Pyrazoles/pharmacokinetics , Triazoles/pharmacokinetics , Adult , Drug Interactions , Erythropoietin/pharmacokinetics , Glucuronosyltransferase/antagonists & inhibitors , Humans , Male , Middle Aged , Young Adult
3.
Basic Clin Pharmacol Toxicol ; 127(3): 221-233, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32248614

ABSTRACT

The absorption, distribution, metabolism and excretion of molidustat were investigated in healthy male participants. In study 1, a mass balance study, radiolabelled molidustat 25 mg (3.57 MBq) was administered as an oral solution (n = 4). Following rapid absorption, molidustat-related radioactivity was predominantly distributed in plasma rather than in red blood cells. The total recovery of the administered radioactivity was 97.0%, which was mainly excreted renally (90.7%). Metabolite M-1, produced by N-glucuronidation, was the dominant component in plasma (80.2% of the area under the concentration-time curve for total radioactivity) and was primarily excreted via urine (~85% of dose). Only minor amounts of unchanged molidustat were excreted in urine (~4%) and faeces (~6%). Study 2 investigated the absolute bioavailability and pharmacodynamics of molidustat (part 1, n = 12; part 2, n = 16). Orally administered molidustat immediate release tablets had an absolute bioavailability of 59%. Following intravenous administration (1, 5 and 25 mg), total body clearance of molidustat was 28.7-34.5 L/h and volume of distribution at steady state was 39.3-50.0 L. All doses of molidustat transiently elevated endogenous erythropoietin levels, irrespective of the route of administration. Molidustat was considered safe and well tolerated at the administered doses.


Subject(s)
Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Triazoles/metabolism , Triazoles/pharmacokinetics , Administration, Oral , Adult , Biological Availability , Drug Evaluation, Preclinical , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Middle Aged , Pyrazoles/blood , Pyrazoles/urine , Tissue Distribution , Triazoles/blood , Triazoles/urine
4.
Eur J Clin Pharmacol ; 76(2): 185-197, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31919558

ABSTRACT

PURPOSE: The present studies assessed the drug-drug interaction of molidustat, a hypoxia-inducible factor prolyl hydroxylase inhibitor, with iron and calcium supplements, which are common medications in patients with anaemia due to chronic kidney disease (CKD). METHODS: Forty-two healthy men received molidustat alone (fasted or fed) or combined with oral iron(II) or calcium(II), given immediately before or between 4 h before and 1 h after molidustat in three randomized, open-label, crossover studies (12-15 participants per study). Molidustat AUC and Cmax were assessed as the main pharmacokinetic parameters, and endogenous erythropoietin (EPO) was measured to evaluate pharmacodynamics. RESULTS: Depending on prandial state, concomitant intake of iron(II) reduced molidustat AUC and Cmax by 50-75% and 46-84%, respectively, and EPO AUC(0-24) and Cmax by 31-44% and 36-48%, respectively. The influence of iron(II) declined with increasing the time interval to the intake of molidustat, with reductions in molidustat AUC and Cmax of 9% and 10%, respectively, when iron(II) intake occurred 4 h before molidustat. Accordingly, effects on endogenous EPO were less pronounced with increased time separation between oral iron(II) and molidustat intake. Calcium(II) reduced molidustat AUC and Cmax by 15% and 47%, respectively, without influence on EPO response. All treatments were well tolerated. CONCLUSIONS: In contrast to concomitant oral intake of calcium, the effect of oral iron supplements on molidustat pharmacokinetics and pharmacodynamics should be considered, and the two agents should be administered with an appropriate time separation.


Subject(s)
Calcium/administration & dosage , Iron/administration & dosage , Pyrazoles/administration & dosage , Triazoles/administration & dosage , Administration, Oral , Adult , Anemia/drug therapy , Area Under Curve , Calcium/pharmacology , Cross-Over Studies , Dietary Supplements , Drug Administration Schedule , Drug Interactions , Erythropoietin/metabolism , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Iron/pharmacology , Male , Middle Aged , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Time Factors , Triazoles/pharmacokinetics , Triazoles/pharmacology , Young Adult
5.
Expert Opin Drug Metab Toxicol ; 15(11): 975-984, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31619082

ABSTRACT

Objectives: Riociguat is a soluble guanylate cyclase stimulator licensed for the treatment of pulmonary arterial hypertension (PAH), a potentially fatal complication of human immunodeficiency virus infection. This study investigated the inhibitory potency of selected antiretroviral regimens on the metabolic clearance of riociguat.Methods: The inhibitory potential of the components of six antiretroviral combinations (ATRIPLA® (efavirenz/emtricitabine/tenofovir disoproxil), COMPLERA® (rilpivirine/emtricitabine/tenofovir disoproxil), STRIBILD® (elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil), TRIUMEQ® (abacavir/dolutegravir/lamivudine), and two ritonavir-boosted regimens) on riociguat metabolism were evaluated in recombinant human CYP1A1 and CYP3A4 as well as in human hepatocytes exhibiting both CYP1A1 and CYP3A4 activity. In vitro-in vivo correlation was performed between calculated and observed increases in riociguat exposure in vivo.Results: Using both in vitro systems, the predicted increase in exposure of riociguat was highest with components of TRIUMEQ® followed by COMPLERA®, ATRIPLA®, STRIBILD®, and the ritonavir-boosted regimens. Further experiments in human hepatocytes confirmed CYP1A1 to be the predominant enzyme in the metabolic clearance of riociguat.Conclusion: Antiretroviral treatment containing the potent CYP1A1 inhibitor abacavir had the greatest impact on riociguat metabolic clearance. The impact of comedications containing only strong CYP3A4 inhibitors e.g. ritonavir was less pronounced, suggesting a benefit of riociguat over PAH-targeting medications with contraindications for use with strong CYP3A4 inhibitors.


Subject(s)
Anti-HIV Agents/pharmacology , Cytochrome P-450 CYP1A1/metabolism , Enzyme Activators/metabolism , Pyrazoles/metabolism , Pyrimidines/metabolism , Anti-HIV Agents/administration & dosage , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions , Enzyme Activators/administration & dosage , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , In Vitro Techniques , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage
6.
Pulm Circ ; 9(2): 2045894019848644, 2019.
Article in English | MEDLINE | ID: mdl-30997864

ABSTRACT

Riociguat, a first-in-class soluble guanylate cyclase stimulator, is approved for the treatment of pulmonary arterial hypertension (PAH), a serious potential complication of human immunodeficiency virus (HIV) infection. This open-label study investigated the pharmacokinetic drug-drug interaction potential of antiretroviral therapies on riociguat exposure in HIV-infected adults. HIV-infected adults without PAH on stable antiretroviral regimens (efavirenz/emtricitabine/tenofovir disoproxil, emtricitabine/rilpivirine/tenofovir disoproxil, elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil, abacavir/dolutegravir/lamivudine, or a ritonavir-boosted triple regimen) for ≥ 6 weeks received a single riociguat dose (0.5 mg). Riociguat pharmacokinetics and safety were assessed; pharmacokinetics was compared with historical healthy volunteer data. Of 41 participants treated (n = 8 in each arm, except n = 9 in the ritonavir-boosted triple regimen arm), 40 were included in the pharmacokinetic analyses. Riociguat median tmax was 1.00-1.27 h, with comparable maximum concentration (Cmax) across the five background antiretroviral groups. Riociguat exposure was highest with abacavir/dolutegravir/lamivudine, followed by elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil > emtricitabine/rilpivirine/tenofovir disoproxil > ritonavir-boosted triple regimen > efavirenz/emtricitabine/tenofovir disoproxil; riociguat area under the plasma concentration versus time curve (AUC) was approximately threefold higher with abacavir/dolutegravir/lamivudine than efavirenz/emtricitabine/tenofovir disoproxil. Compared with historical data, riociguat exposure in HIV-infected adults was similar when co-administered with efavirenz/emtricitabine/tenofovir disoproxil, slightly increased when administered with ritonavir-boosted triple regimen and increased by approximately threefold when administered with abacavir/dolutegravir/lamivudine. Riociguat was well tolerated, with no new safety findings. Riociguat was well tolerated in adults with HIV on stable background antiretroviral therapy although an apparent increase in AUC of riociguat was observed in patients receiving abacavir/dolutegravir/lamivudine. Patients should be monitored closely during riociguat initiation and dose adjustment for signs and symptoms of hypotension.

7.
Pulm Pharmacol Ther ; 56: 86-93, 2019 06.
Article in English | MEDLINE | ID: mdl-30917927

ABSTRACT

BACKGROUND: There are only limited treatment options for patients with non-cystic fibrosis bronchiectasis (non-CF BE). Human neutrophil elastase (HNE) is a mediator of tissue destruction in non-CF BE. BAY 85-8501, a selective and reversible HNE inhibitor, could represent a new treatment option for this disease. METHODS: This was a phase 2a, randomized, placebo-controlled, double-blind, parallel-group study. The primary objective was to assess the safety and tolerability of 1 mg BAY 85-8501 once daily (OD) for 28 days compared with placebo in patients with non-CF BE. Secondary objectives were to investigate the effects of 4 weeks of treatment with BAY 85-8501 on health-related quality of life, pulmonary function, and inflammatory and tissue damage biomarkers in sputum, blood and/or urine, and to evaluate the pharmacokinetics of BAY 85-8501. RESULTS: Overall, 94 patients (mean age, 66 years; 53% male) were randomized (n = 47 per group), and 82 completed the study (BAY 85-8501, n = 37; placebo, n = 45). Treatment-emergent adverse events (TEAEs) occurred in 31 patients (66%) taking BAY 85-8501 and in 36 patients (77%) taking placebo, and were mostly mild or moderate. The serious TEAEs (BAY 85-8501, n = 3; placebo, n = 1) were not considered to be study-drug related. There were no changes in pulmonary function parameters from baseline to end of treatment, and health-related quality of life did not improve in any group. HNE activity in blood after zymosan challenge decreased significantly with BAY 85-8501 treatment (P = 0.0250 versus placebo). There were no significant differences in other biomarkers between treatment groups, with the exception of a small increase in interleukin-8 levels in sputum in the BAY 85-8501 group. Trough plasma concentrations of BAY 85-8501 plateaued after 2 weeks. CONCLUSIONS: 1 mg BAY 85-8501 OD had a favourable safety and tolerability profile when administered for 28 days to patients with non-CF BE. Further studies with a longer treatment duration are needed to evaluate the potential clinical efficacy in this study population.


Subject(s)
Bronchiectasis/drug therapy , Leukocyte Elastase/antagonists & inhibitors , Proteinase Inhibitory Proteins, Secretory/therapeutic use , Pyrimidinones/therapeutic use , Sulfones/therapeutic use , Aged , Bronchiectasis/physiopathology , Double-Blind Method , Female , Humans , Male , Middle Aged , Proteinase Inhibitory Proteins, Secretory/adverse effects , Proteinase Inhibitory Proteins, Secretory/pharmacokinetics , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics , Quality of Life , Sputum/metabolism , Sulfones/adverse effects , Sulfones/pharmacokinetics , Treatment Outcome
8.
Eur J Pharm Sci ; 125: 142-150, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30273661

ABSTRACT

PURPOSE: Optimize adult fecal material composition for evaluating the clinical importance of bacterial degradation of therapeutic agents in the lower intestine (distal small intestine, D-SI and proximal colon, P-COL). Evaluate the usefulness of optimized fecal material in the evaluation of bacterial degradation of five model highly permeable drugs: two nitroreductase substrates (nitrendipine and nimodipine), three drugs for which published data indicate no impact of bacterial degradation on in vivo performance (levodopa, budesonide and rivaroxaban) and one prodrug (sulfasalazine, an azoreductase substrate) from which a locally acting on the mucosa of the lower intestine drug is derived (mesalamine). METHODS: 30 min and 95 min were used as point estimates of maximum bacterial degradation half-lives for bacterial degradation in D-SI or in P-COL, respectively, to be clinically important, i.e. for at least 20% reduction in absorption from D-SI or P-COL to occur. Optimization of fecal material was based on recently reported degradation profiles of metronidazole (a nitroreductase substrate) and olsalazine (an azoreductase substrate) in the lower intestine of healthy adults which are clinically important. Model compounds were tested in optimized fecal materials and data were evaluated vs. existing in vivo data in adults. RESULTS: Simulated ileal bacteria (SIB) consisted of 5.5% (w/v) stools in normal saline and simulated colonic bacteria (SCoB) consisted of 8.3% (w/v) stools in normal saline. For all model compounds, data in SIB and SCoB were in line with available information in adults. [Degradation half-life in SIB/Degradation half-life in SCoB] ≈ [Stool content in SCoB/Stool content in SIB] ≈ 1.5, i.e. bacterial degradation in SIB could be predicted from bacterial degradation in SCoB. CONCLUSION: Data in SCoB only are useful for evaluating whether bacterial degradation in P-COL and in D-SI is likely to be clinically important for orally administered, highly permeable drugs or prodrugs which act locally after bacterial degradation. The usefulness of this approach in cases where enzymes other than nitroreductases or azoreductases are involved requires further confirmation.


Subject(s)
Bacteria/metabolism , Feces/chemistry , Intestines/microbiology , Pharmaceutical Preparations/metabolism , Adult , Gastrointestinal Microbiome , Humans , Male , Middle Aged
9.
Clin Pharmacokinet ; 57(6): 647-661, 2018 06.
Article in English | MEDLINE | ID: mdl-29086344

ABSTRACT

Oral riociguat is a soluble guanylate cyclase (sGC) stimulator that targets the nitric oxide (NO)-sGC-cyclic guanosine monophosphate pathway with a dual mode of action: directly by stimulating sGC, and indirectly by increasing the sensitivity of sGC to NO. It is rapidly absorbed, displays almost complete bioavailability (94.3%), and can be taken with or without food and as crushed or whole tablets. Riociguat exposure shows pronounced interindividual (60%) and low intraindividual (30%) variability in patients with pulmonary arterial hypertension (PAH) or chronic thromboembolic pulmonary hypertension (CTEPH), and is therefore administered using an individual dose-adjustment scheme at treatment initiation. The half-life of riociguat is approximately 12 h in patients and approximately 7 h in healthy individuals. Riociguat and its metabolites are excreted via both renal (33-45%) and biliary routes (48-59%), and dose adjustment should be performed with particular care in patients with moderate hepatic impairment or mild to severe renal impairment (no data exist for patients with severe hepatic impairment). The pharmacodynamic effects of riociguat reflect the action of a vasodilatory agent, and the hemodynamic response to riociguat correlated with riociguat exposure in patients with PAH or CTEPH in phase III population pharmacokinetic/pharmacodynamic analyses. Riociguat has a low risk of clinically relevant drug interactions due to its clearance by multiple cytochrome P450 (CYP) enzymes and its lack of effect on major CYP isoforms and transporter proteins at therapeutic levels. Riociguat has been approved for the treatment of PAH and CTEPH that is inoperable or persistent/recurrent after surgical treatment.


Subject(s)
Pyrazoles/pharmacology , Pyrimidines/pharmacology , Administration, Oral , Animals , Drug Interactions , Humans , Hypertension, Pulmonary/metabolism , Pyrazoles/blood , Pyrazoles/pharmacokinetics , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Soluble Guanylyl Cyclase
10.
Cancer Chemother Pharmacol ; 80(3): 535-544, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714036

ABSTRACT

PURPOSE: To determine the pharmacokinetics of radiolabeled copanlisib (BAY 80-6946) in healthy male volunteers and to investigate the disposition and biotransformation of copanlisib. METHODS: A single dose of 12 mg copanlisib containing 2.76 MBq [14C]copanlisib was administered as a 1-h intravenous infusion to 6 volunteers with subsequent sampling up to 34 days. Blood, plasma, urine and feces were collected to monitor total radioactivity, parent compound and metabolites. RESULTS: Copanlisib treatment was well tolerated. Copanlisib was rapidly distributed throughout the body with a volume distribution of 1870 L and an elimination half-life of 52.1-h (range 40.4-67.5-h). Copanlisib was the predominant component in human plasma (84% of total radioactivity AUC) and the morpholinone metabolite M1 was the only circulating metabolite (about 5%). Excretion of drug-derived radioactivity based on all 6 subjects was 86% of the dose within a collection interval of 20-34 days with 64% excreted into feces as major route of elimination and 22% into urine. Unchanged copanlisib was the main component excreted into urine (15% of dose) and feces (30% of dose). Excreted metabolites (41% of dose) of copanlisib resulted from oxidative biotransformation. CONCLUSIONS: Copanlisib was eliminated predominantly in the feces compared to urine as well as by hepatic biotransformation, suggesting that the clearance of copanlisib would more likely be affected by hepatic impairment than by renal dysfunction. The dual mode of elimination via unchanged excretion of copanlisib and oxidative metabolism decreases the risk of clinically relevant PK-related drug-drug interactions.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Quinazolines/therapeutic use , Administration, Intravenous , Class I Phosphatidylinositol 3-Kinases/pharmacokinetics , Class I Phosphatidylinositol 3-Kinases/therapeutic use , Healthy Volunteers , Humans , Male , Middle Aged , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics
11.
Eur J Pharm Sci ; 96: 598-609, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27671970

ABSTRACT

Predicting oral bioavailability (Foral) is of importance for estimating systemic exposure of orally administered drugs. Physiologically-based pharmacokinetic (PBPK) modelling and simulation have been applied extensively in biopharmaceutics recently. The Oral Biopharmaceutical Tools (OrBiTo) project (Innovative Medicines Initiative) aims to develop and improve upon biopharmaceutical tools, including PBPK absorption models. A large-scale evaluation of PBPK models may be considered the first step. Here we characterise the OrBiTo active pharmaceutical ingredient (API) database for use in a large-scale simulation study. The OrBiTo database comprised 83 APIs and 1475 study arms. The database displayed a median logP of 3.60 (2.40-4.58), human blood-to-plasma ratio of 0.62 (0.57-0.71), and fraction unbound in plasma of 0.05 (0.01-0.17). The database mainly consisted of basic compounds (48.19%) and Biopharmaceutics Classification System class II compounds (55.81%). Median human intravenous clearance was 16.9L/h (interquartile range: 11.6-43.6L/h; n=23), volume of distribution was 80.8L (54.5-239L; n=23). The majority of oral formulations were immediate release (IR: 87.6%). Human Foral displayed a median of 0.415 (0.203-0.724; n=22) for IR formulations. The OrBiTo database was found to be largely representative of previously published datasets. 43 of the APIs were found to satisfy the minimum inclusion criteria for the simulation exercise, and many of these have significant gaps of other key parameters, which could potentially impact the interpretability of the simulation outcome. However, the OrBiTo simulation exercise represents a unique opportunity to perform a large-scale evaluation of the PBPK approach to predicting oral biopharmaceutics.


Subject(s)
Biopharmaceutics/methods , Databases, Factual , Models, Biological , Pharmaceutical Preparations/metabolism , Administration, Oral , Drug Evaluation, Preclinical/methods , Forecasting , Humans , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Pharmaceutical Preparations/administration & dosage
12.
Eur J Pharm Biopharm ; 105: 193-202, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27322002

ABSTRACT

AIMS: Food intake is known to have various effects on gastrointestinal luminal conditions in terms of transit times, hydrodynamic forces and/or luminal fluid composition and can therefore affect the dissolution behavior of solid oral dosage forms. The aim of this study was to investigate and detect the dosage form-dependent food effect that has been observed for two extended-release formulations of nifedipine using in vitro dissolution tests. METHODS: Two monolithic extended release formulations, the osmotic pump Adalat® XL 60mg and matrix-type Adalat® Eins 30mg formulation, were investigated with biorelevant dissolution methods using the USP apparatus III and IV under both simulated prandial states, and their corresponding quality control dissolution method. In vitro data were compared to published and unpublished in vivo data using deconvolution-based in vitro - in vivo correlation (IVIVC) approaches. RESULTS: Quality control dissolution methods tended to overestimate the dissolution rate due to the excessive solubilizing capabilities of the sodium dodecyl sulfate (SDS)-containing dissolution media. Using Level II biorelevant media the dosage form dependent food effect for nifedipine was described well when studied with the USP apparatus III, whereas the USP apparatus IV failed to detect the positive food effect for the matrix-type dosage form. CONCLUSIONS: It was demonstrated that biorelevant methods can serve as a useful tool during formulation development as they were able to qualitatively reflect the in vivo data.


Subject(s)
Calcium Channel Blockers/pharmacology , Dosage Forms , Food-Drug Interactions , Nifedipine/pharmacology , Calcium Channel Blockers/administration & dosage , Delayed-Action Preparations , Humans , Nifedipine/administration & dosage , Solubility
13.
Pulm Circ ; 6(Suppl 1): S97-S102, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27162633

ABSTRACT

Female patients requiring treatment for pulmonary arterial hypertension (PAH) are advised to avoid pregnancy because of the high associated mortality rate. Oral contraception is one of the main methods of preventing pregnancy in this context, mandating pharmacokinetic and safety studies for new agents in this setting. Riociguat is a soluble guanylate cyclase stimulator approved for treatment of PAH and inoperable and persistent or recurrent chronic thromboembolic pulmonary hypertension. This single-center, randomized, nonblinded study involving healthy postmenopausal women investigated the effect of riociguat on plasma concentrations of levonorgestrel (0.15 mg) and ethinylestradiol (0.03 mg) in a combined oral contraceptive. Treatment A was a single oral tablet of levonorgestrel-ethinylestradiol. In treatment B, subjects received 2.5 mg riociguat 3 times daily for 12 days. On the eighth day, they also received a single oral tablet of levonorgestrel-ethinylestradiol. Subjects received both regimens in a crossover design. There was no change in area under the plasma concentration-time curves of levonorgestrel or ethinylestradiol or maximum concentration in plasma (C max) of levonorgestrel during combined administration versus levonorgestrel-ethinylestradiol alone. A 20% increase in the C max of ethinylestradiol was noted during coadministration; this is not anticipated to adversely impact the contraceptive efficacy or to require any dose adjustment for ethinylestradiol. Plasma concentrations and exposures of riociguat were within the expected range and were not influenced by coadministration with levonorgestrel-ethinylestradiol. Combined treatment was safe and well tolerated. In conclusion, riociguat did not change the exposure to levonorgestrel or ethinylestradiol relative to oral contraceptive administered alone.

14.
Eur J Pharm Biopharm ; 102: 191-201, 2016 May.
Article in English | MEDLINE | ID: mdl-26955751

ABSTRACT

The purpose of the study was to experimentally deduce pH-dependent critical volumes to dissolve applied dose (VDAD) that determine whether a drug candidate can be developed as immediate release (IR) tablet containing crystalline API, or if solubilization technology is needed to allow for sufficient oral bioavailability. pH-dependent VDADs of 22 and 83 compounds were plotted vs. the relative oral bioavailability (AUC solid vs. AUC solution formulation, Frel) in humans and rats, respectively. Furthermore, in order to investigate to what extent Frel rat may predict issues with solubility limited absorption in human, Frel rat was plotted vs. Frel human. Additionally, the impact of bile salts and lecithin on in vitro dissolution of poorly soluble compounds was tested and data compared to Frel rat and human. Respective in vitro - in vivo and in vivo - in vivo correlations were generated and used to build developability criteria. As a result, based on pH-dependent VDAD, Frel rat and in vitro dissolution in simulated intestinal fluid the IR formulation strategy within Pharmaceutical Research and Development organizations can be already set at late stage of drug discovery.


Subject(s)
Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Administration, Oral , Animals , Bile Acids and Salts/chemistry , Biological Availability , Biopharmaceutics/methods , Caco-2 Cells , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Humans , Lecithins/chemistry , Permeability , Pharmaceutical Preparations/metabolism , Rats , Solubility , Tablets/administration & dosage , Tablets/chemistry , Tablets/pharmacokinetics
15.
J Med Chem ; 57(15): 6739-50, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25051097

ABSTRACT

Bivalent ligands of G protein-coupled receptors have been shown to simultaneously either bind to two adjacent receptors or to bridge different parts of one receptor protein. Recently, we found that bivalent agonists of muscarinic receptors can simultaneously occupy both the orthosteric transmitter binding site and the allosteric vestibule of the receptor protein. Such dualsteric agonists display a certain extent of subtype selectivity, generate pathway-specific signaling, and in addition may allow for designed partial agonism. Here, we want to extend the concept to bivalent antagonism. Using the phthal- and naphthalimide moieties, which bind to the allosteric, extracellular site, and atropine or scopolamine as orthosteric building blocks, both connected by a hexamethonium linker, we were able to prove a bitopic binding mode of antagonist hybrids for the first time. This is demonstrated by structure-activity relationships, site-directed mutagenesis, molecular docking studies, and molecular dynamics simulations. Findings revealed that a difference in spatial orientation of the orthosteric tropane moiety translates into a divergent M2/M5 subtype selectivity of the corresponding bitopic hybrids.


Subject(s)
Atropine Derivatives/chemistry , Muscarinic Antagonists/chemistry , Naphthalimides/chemistry , Phthalimides/chemistry , Scopolamine Derivatives/chemistry , Allosteric Regulation , Animals , Atropine Derivatives/chemical synthesis , Atropine Derivatives/pharmacology , Binding Sites , CHO Cells , Cricetulus , Drug Inverse Agonism , Humans , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Muscarinic Agonists/chemical synthesis , Muscarinic Agonists/chemistry , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/chemical synthesis , Muscarinic Antagonists/pharmacology , Mutation , Naphthalimides/chemical synthesis , Naphthalimides/pharmacology , Phthalimides/chemical synthesis , Phthalimides/pharmacology , Radioligand Assay , Receptor, Muscarinic M2/agonists , Receptor, Muscarinic M2/antagonists & inhibitors , Receptor, Muscarinic M2/genetics , Scopolamine Derivatives/chemical synthesis , Scopolamine Derivatives/pharmacology , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...