Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Clin Pharmacol Drug Dev ; 13(6): 590-600, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38623935

ABSTRACT

This open-label, phase 1 study was conducted with healthy adult participants to evaluate the potential drug-drug interaction between rilzabrutinib and quinidine (an inhibitor of P-glycoprotein [P-gp] and CYP2D6) or rifampin (an inducer of CYP3A and P-gp). Plasma concentrations of rilzabrutinib were measured after a single oral dose of rilzabrutinib 400 mg administered on day 1 and again, following a wash-out period, after co-administration of rilzabrutinib and quinidine or rifampin. Specifically, quinidine was given at a dose of 300 mg every 8 hours for 5 days from day 7 to day 11 (N = 16) while rifampin was given as 600 mg once daily for 11 days from day 7 to day 17 (N = 16) with rilzabrutinib given in the morning of day 10 (during quinidine dosing) or day 16 (during rifampin dosing). Quinidine had no significant effect on rilzabrutinib pharmacokinetics. Rifampin decreased rilzabrutinib exposure (the geometric mean of Cmax and AUC0-∞ decreased by 80.5% and 79.5%, respectively). Single oral doses of rilzabrutinib, with or without quinidine or rifampin, appeared to be well tolerated. These findings indicate that rilzabrutinib is a substrate for CYP3A but not a substrate for P-gp.


Subject(s)
Area Under Curve , Drug Interactions , Healthy Volunteers , Quinidine , Rifampin , Humans , Rifampin/administration & dosage , Rifampin/adverse effects , Quinidine/administration & dosage , Quinidine/adverse effects , Quinidine/pharmacology , Quinidine/pharmacokinetics , Adult , Male , Female , Young Adult , Middle Aged , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A Inducers/adverse effects , Cytochrome P-450 CYP3A/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Pyrimidines/adverse effects
2.
CPT Pharmacometrics Syst Pharmacol ; 13(4): 544-550, 2024 04.
Article in English | MEDLINE | ID: mdl-38343040

ABSTRACT

Personalized dosing approaches play important roles in clinical practices to improve benefit: risk profiles. Whereas this is also important for drug development, especially in the context of drugs with narrow therapeutic windows, such approaches have not been fully evaluated during clinical development. Fazpilodemab (BFKB8488A) is an agonistic bispecific antibody which was being developed for the treatment of nonalcoholic steatohepatitis. The objective of this study was to characterize the exposure-response relationships of fazpilodemab with the purpose of guiding dose selection for a phase II study, as well as to evaluate various personalized dosing strategies to optimize the treatment benefit. Fazpilodemab exhibited clear exposure-response relationships for a pharmacodynamic (PD) biomarker and gastrointestinal adverse events (GIAEs), such as nausea and vomiting. Static exposure-response analysis, as well as longitudinal adverse event (AE) analysis using discrete-time Markov model, were performed to characterize the observations. Clinical trial simulations were performed based on the developed exposure-response models to evaluate probability of achieving target PD response and the frequency of GIAEs to inform phase II dose selection. Dynamic simulation of personalized dosing strategies demonstrated that the AE-based personalized dosing is the most effective approach for optimizing the benefit-risk profiles. The approach presented here can be a useful framework for quantifying the benefit of personalized dosing for drugs with narrow therapeutic windows.


Subject(s)
Klotho Proteins , Humans
3.
Nat Med ; 27(11): 1961-1969, 2021 11.
Article in English | MEDLINE | ID: mdl-34750553

ABSTRACT

Bruton's tyrosine kinase (BTK) is crucial for FcεRI-mediated mast cell activation and essential for autoantibody production by B cells in chronic spontaneous urticaria (CSU). Fenebrutinib, an orally administered, potent, highly selective, reversible BTK inhibitor, may be effective in CSU. This double-blind, placebo-controlled, phase 2 trial (EudraCT ID 2016-004624-35 ) randomized 93 adults with antihistamine-refractory CSU to 50 mg daily, 150 mg daily and 200 mg twice daily of fenebrutinib or placebo for 8 weeks. The primary end point was change from baseline in urticaria activity score over 7 d (UAS7) at week 8. Secondary end points were the change from baseline in UAS7 at week 4 and the proportion of patients well-controlled (UAS7 ≤ 6) at week 8. Fenebrutinib efficacy in patients with type IIb autoimmunity and effects on IgG-anti-FcεRI were exploratory end points. Safety was also evaluated. The primary end point was met, with dose-dependent improvements in UAS7 at week 8 occurring at 200 mg twice daily and 150 mg daily, but not at 50 mg daily of fenebrutinib versus placebo. Asymptomatic, reversible grade 2 and 3 liver transaminase elevations occurred in the fenebrutinib 150 mg daily and 200 mg twice daily groups (2 patients each). Fenebrutinib diminished disease activity in patients with antihistamine-refractory CSU, including more patients with refractory type IIb autoimmunity. These results support the potential use of BTK inhibition in antihistamine-refractory CSU.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Chronic Urticaria/drug therapy , Histamine H1 Antagonists/therapeutic use , Histamine Release/drug effects , Piperazines/therapeutic use , Pyridones/therapeutic use , Adolescent , Adult , Angioedema/drug therapy , Autoimmunity/immunology , Double-Blind Method , Drug Resistance/physiology , Female , Histamine H1 Antagonists/adverse effects , Humans , Immunoglobulin E/immunology , Male , Mast Cells/metabolism , Middle Aged , Piperazines/adverse effects , Placebos/administration & dosage , Pyridones/adverse effects , Receptors, IgE/antagonists & inhibitors , Transaminases/analysis , Young Adult
4.
Arthritis Rheumatol ; 73(10): 1835-1846, 2021 10.
Article in English | MEDLINE | ID: mdl-34042314

ABSTRACT

OBJECTIVE: Fenebrutinib (GDC-0853) is a noncovalent, oral, and highly selective inhibitor of Bruton's tyrosine kinase (BTK). The efficacy, safety, and pharmacodynamics of fenebrutinib in systemic lupus erythematosus (SLE) were assessed in this phase II, multicenter, randomized, placebo-controlled study. METHODS: Patients who had moderately to severely active SLE while receiving background standard therapy were randomized to receive placebo, fenebrutinib 150 mg once daily, or fenebrutinib 200 mg twice daily. Glucocorticoid taper was recommended from weeks 0 to 12 and from weeks 24 to 36. The primary end point was the SLE Responder Index 4 (SRI-4) response at week 48. RESULTS: Patients (n = 260) were enrolled from 44 sites in 12 countries, with the majority from Latin America, the US, and Western Europe. The SRI-4 response rates at week 48 were 51% for fenebrutinib 150 mg once daily (P = 0.37 versus placebo), 52% for fenebrutinib 200 mg twice daily (P = 0.34 versus placebo), and 44% for placebo. British Isles Lupus Assessment Group-based Combined Lupus Assessment response rates at week 48 were 53% for fenebrutinib 150 mg once daily (P = 0.086 versus placebo), 42% for fenebrutinib 200 mg twice daily (P = 0.879 versus placebo), and 41% for placebo. Safety results were similar across all arms, although serious adverse events were more frequent with fenebrutinib 200 mg twice daily. By week 48, patients treated with fenebrutinib had reduced levels of a BTK-dependent plasmablast RNA signature, anti-double-stranded DNA autoantibodies, total IgG, and IgM, as well as increased complement C4 levels, all relative to placebo. CONCLUSION: While fenebrutinib had an acceptable safety profile, the primary end point, SRI-4 response, was not met despite evidence of strong pathway inhibition.


Subject(s)
Antirheumatic Agents/therapeutic use , Autoantibodies/blood , Lupus Erythematosus, Systemic/drug therapy , Piperazines/therapeutic use , Pyridones/therapeutic use , Adolescent , Adult , Aged , Antirheumatic Agents/adverse effects , Antirheumatic Agents/pharmacology , Complement C3/metabolism , Complement C4/metabolism , Double-Blind Method , Female , Humans , Lupus Erythematosus, Systemic/blood , Male , Middle Aged , Piperazines/adverse effects , Piperazines/pharmacology , Pyridones/adverse effects , Pyridones/pharmacology , Treatment Outcome , Young Adult
5.
Proc Natl Acad Sci U S A ; 117(46): 28992-29000, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33139537

ABSTRACT

Fibroblast growth factor 21 (FGF21) controls metabolic organ homeostasis and eating/drinking behavior via FGF receptor 1/Klothoß (FGFR1/KLB) complexes expressed in adipocytes, pancreatic acinar cells, and the nervous system in mice. Chronic administration of recombinant FGF21 or engineered variants improves metabolic health in rodents, nonhuman primates, and humans; however, the rapid turnover of these molecules limits therapeutic utility. Here we show that the bispecific anti-FGFR1/KLB agonist antibody BFKB8488A induced marked weight loss in obese cynomolgus monkeys while elevating serum adiponectin and the adipose expression of FGFR1 target genes, demonstrating its action as an FGF21 mimetic. In a randomized, placebo-controlled, single ascending-dose study in overweight/obese human participants, subcutaneous BFKB8488A injection caused transient body weight reduction, sustained improvement in cardiometabolic parameters, and a trend toward reduction in preference for sweet taste and carbohydrate intake. These data suggest that specific activation of the FGFR1/KLB complex in humans can be used as therapy for obesity-related metabolic defects.


Subject(s)
Food Preferences , Obesity/drug therapy , Obesity/metabolism , Receptor, Fibroblast Growth Factor, Type 1/immunology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adiponectin/blood , Adipose Tissue/metabolism , Adolescent , Adult , Aged , Animals , Antibodies/therapeutic use , Biomarkers/blood , Body Weight , Female , Fibroblast Growth Factors , Homeostasis , Humans , Macaca fascicularis , Male , Mice , Middle Aged , Weight Loss , Young Adult
6.
Clin Pharmacol Ther ; 108(6): 1224-1232, 2020 12.
Article in English | MEDLINE | ID: mdl-32535897

ABSTRACT

Cyclodextrins are widely used pharmaceutical excipients, particularly for insoluble compounds dosed orally, such as the oral solution of itraconazole, which is frequently used in clinical drug-drug interaction studies to inhibit cytochrome P450 3A. Since cyclodextrins act by forming inclusion complexes with their coformulated drug, they could have an unintended consequence of affecting absorption if they form a strong complex with the potential victim drug in an itraconazole drug-drug interaction study. This observation was made in a drug-drug interaction study with the Bruton's tyrosine kinase (BTK) inhibitor fenebrutinib and itraconazole, in which, relative to the control group, the expected increase in fenebrutinib maximum plasma concentration (Cmax ) was not observed in the itraconazole group, and a delay in time to reach maximum plasma concentration (Tmax ) was observed in the itraconazole group. The in vitro binding constant between fenebrutinib and hydroxypropyl-ß-cyclodextrin was determined to be 2 × 105  M-1 , and the apparent permeability of fenebrutinib across a Madin-Darby canine kidney cell monolayer decreased in a cyclodextrin concentration-dependent manner. This observation was confirmed in vivo, in a pentagastrin-pretreated dog model, in which fenebrutinib was administered with or without cyclodextrin; a reduction in Cmax , a prolonged Tmax , and increased fenebrutinib recovery in feces replicated the previous observation in healthy volunteers and supported the hypothesis that complexation with cyclodextrin decreased rate and extent of fenebrutinib absorption. Physiologically-based pharmacokinetic modeling was used to translate the in vitro effect of cyclodextrin on fenebrutinib apparent permeability to the in vivo effect on absorption, which was then confirmed using the in vivo dog pharmacokinetic data.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/adverse effects , Excipients/administration & dosage , Intestinal Absorption/drug effects , Itraconazole/adverse effects , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyridones/pharmacokinetics , 2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Administration, Oral , Adolescent , Adult , Animals , Dogs , Drug Administration Schedule , Drug Interactions , Excipients/toxicity , Feces/chemistry , Female , Humans , Itraconazole/administration & dosage , Madin Darby Canine Kidney Cells , Male , Middle Aged , Models, Animal , Permeability , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridones/administration & dosage , Young Adult
7.
CPT Pharmacometrics Syst Pharmacol ; 9(6): 332-341, 2020 06.
Article in English | MEDLINE | ID: mdl-32383787

ABSTRACT

Fenebrutinib is a CYP3A substrate and time-dependent inhibitor, as well as a BCRP and OATP1B transporter inhibitor in vitro. Physiologically-based pharmacokinetic (PBPK) modeling strategies with the ultimate goal of understanding complex drug-drug interactions (DDIs) and proposing doses for untested scenarios were developed. The consistency in the results of two independent approaches, PBPK simulation and endogenous biomarker measurement, supported that the observed transporter DDI is primarily due to fenebrutinib inhibition of intestinal BCRP, rather than hepatic OATP1B. A mechanistic-absorption model accounting for the effects of excipient complexation with fenebrutinib was used to rationalize the unexpected observation of itraconazole-fenebrutinib DDI (maximum plasma concentration (Cmax ) decreased, and area under the curve (AUC) increased). The totality of the evidence from sensitivity analysis and clinical and nonclinical data suggested that fenebrutinib is likely a sensitive CYP3A substrate. This advanced PBPK application allowed the use of model-informed approach to facilitate the development of concomitant medication recommendations for fenebrutinib without requiring additional clinical DDI studies.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Intestines/drug effects , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Liver/drug effects , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Piperazines/pharmacokinetics , Pyridones/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Biotransformation , Clinical Trials, Phase I as Topic , Computer Simulation , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Cytochrome P-450 CYP3A Inhibitors/chemistry , Dogs , Drug Compounding , Drug Development , Drug Interactions , Excipients/chemistry , Humans , Liver/metabolism , Liver-Specific Organic Anion Transporter 1/metabolism , Madin Darby Canine Kidney Cells , Neoplasm Proteins/metabolism , Piperazines/adverse effects , Piperazines/chemistry , Pyridones/adverse effects , Pyridones/chemistry , Retrospective Studies
8.
Arthritis Rheumatol ; 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32270926

ABSTRACT

OBJECTIVE: To evaluate fenebrutinib, an oral and highly selective non-covalent inhibitor of Bruton's tyrosine kinase (BTK), in patients with active rheumatoid arthritis (RA). METHODS: Patients with RA and inadequate response to methotrexate (cohort 1, n=480) were randomized to fenebrutinib (50 mg once daily, 150 mg once daily, 200 mg twice daily), 40 mg adalimumab every other week, or placebo. Patients with RA and inadequate response to tumor necrosis factor inhibitors (cohort 2, n=98) received fenebrutinib (200 mg twice daily) or placebo. Both cohorts continued methotrexate therapy. RESULTS: In cohort 1, American College of Rheumatology scores (ACR50) at week 12 were similar for fenebrutinib 50 mg once daily and placebo, and higher for fenebrutinib 150 mg once daily (28%) and 200 mg twice daily (35%) than placebo (15%) (p=0.017; p=0.0003). Fenebrutinib 200 mg twice daily and adalimumab (36%) were comparable (p=0.81). In cohort 2, more patients achieved ACR50 with fenebrutinib 200 mg twice daily (25%) than placebo (12%) (p=0.072). The most common adverse events for fenebrutinib included nausea, headache, anemia, and upper respiratory tract infections. Fenebrutinib had significant effects on myeloid and B cell biomarkers (CCL4 and rheumatoid factor). Fenebrutinib and adalimumab caused overlapping as well as distinct changes in B cell and myeloid biomarkers. CONCLUSION: Fenebrutinib demonstrated efficacy comparable to adalimumab in patients with an inadequate response to methotrexate, and safety consistent with existing immunomodulatory therapies for RA. These data support targeting both B and myeloid cells via this novel mechanism for potential efficacy in the treatment of RA.

9.
Pharm Res ; 37(3): 41, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980965

ABSTRACT

The article was published with an incomplete title. The complete title is "Population Pharmacokinetics, Efficacy Exposure-response Analysis, and Model-based Meta-analysis of Fenebrutinib in Subjects with Rheumatoid Arthritis" The original article has been corrected.

10.
Pharm Res ; 37(2): 25, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31907670

ABSTRACT

PURPOSE: Fenebrutinib (GDC-0853), a Bruton's tyrosine kinase (BTK) inhibitor was investigated in a Phase 2 clinical trial in patients with rheumatoid arthritis (RA). Our aim was to apply a model-informed drug development (MIDD) approach to examine the totality of available clinical efficacy data. METHODS: Population pharmacokinetics (popPK) modeling, exposure-response (E-R) analysis, and model-based meta-analysis (MBMA) of fenebrutinib were performed based on the Phase 2 data. RESULTS: PopPK of fenebrutinib after oral administration was described using a 3-compartment model with linear elimination and a flexible absorption transit compartment model. Healthy subjects had a 52% higher apparent clearance than patients. E-R analyses based on longitudinal ACR20, ACR50, and ACR70 and DAS28 (CRP) data modeled fenebrutinib effect with an Emax function, and an efficacy plateau was achieved within the exposure range obtained in the Phase 2 clinical trial. Based on literature data, a summary-level clinical efficacy database was constructed, and MBMA determined ACR20, ACR50, and ACR70 responder rates in the placebo and adalimumab arms of the Phase 2 clinical trial were found to be consistent with historical data for these treatments. CONCLUSIONS: Our multi-pronged approach applied MIDD to maximize knowledge extraction of efficacy data and enabled robust interpretation from a Phase 2 clinical trial.

11.
Clin Pharmacol Ther ; 107(1): 269-277, 2020 01.
Article in English | MEDLINE | ID: mdl-31376152

ABSTRACT

Mechanistic understanding of complex clinical drug-drug interactions (DDIs) with potential involvement of multiple elimination pathways has been challenging, especially given the general lack of specific probe substrates for transporters. Here, we conducted a clinical DDI study to evaluate the interaction potential of fenebrutinib using midazolam (MDZ; CYP3A), simvastatin (CYP3A and OATP1B), and rosuvastatin (BCRP and OATP1B) as probe substrates. Fenebrutinib (200 mg) increased the area under the curve (AUC) of these probe substrates twofold to threefold. To evaluate the mechanism of the observed DDIs, we measured the concentration of coproporphyrin I (CP-I) and coproporphyrin III (CP-III), endogenous biomarkers of OATP1B. There was no change in CP-I or CP-III levels with fenebrutinib, suggesting that the observed DDIs were caused by inhibition of CYP3A and BCRP rather than OATP1B, likely due to increased bioavailability. This is the first published account using an endogenous transporter biomarker to understand the mechanism of complex DDIs involving multiple elimination pathways.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Cytochrome P-450 CYP3A/drug effects , Neoplasm Proteins/antagonists & inhibitors , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adult , Area Under Curve , Biomarkers/metabolism , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Female , Humans , Liver-Specific Organic Anion Transporter 1/drug effects , Liver-Specific Organic Anion Transporter 1/metabolism , Male , Midazolam/pharmacokinetics , Middle Aged , Neoplasm Proteins/metabolism , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridones/administration & dosage , Rosuvastatin Calcium/pharmacokinetics , Simvastatin/pharmacokinetics , Young Adult
12.
J Pharmacol Exp Ther ; 371(1): 202-207, 2019 10.
Article in English | MEDLINE | ID: mdl-31371481

ABSTRACT

Fenebrutinib (GDC-0853) is an orally administered small molecule inhibitor of Bruton's tyrosine kinase being investigated for treatment of rheumatoid arthritis in patients with inadequate responses to methotrexate (MTX). This study interrogated the potential for pharmacokinetic drug interactions between fenebrutinib and MTX. Eighteen healthy male subjects were enrolled in the study. They received a single oral dose of MTX (7.5 mg) on day 1 followed by a 13-day washout period. Subsequently, on days 15-20 the participants received 200 mg of fenebrutinib twice daily. On day 21, they received a 7.5 mg dose of MTX and a 200 mg dose of fenebrutinib under fasting conditions. The geometric mean ratios of MTX area under the plasma concentration-time curve (AUC) and C max on day 21 relative to day 1 (90% confidence interval [CI]) were 0.96 (0.88-1.04) and 1.05 (0.94-1.18), respectively. The geometric mean ratios of fenebrutinib AUC and C max for day 21 relative to day 20 (90% CI) were 1.03 (0.95-1.11) and 1.02 (0.90-1.15), respectively. The combination treatment was well tolerated, with an adverse event profile similar to that reported in other MTX trials. These results indicate that there is no clinically significant pharmacokinetic interaction between fenebrutinib and MTX.


Subject(s)
Antirheumatic Agents/pharmacokinetics , Methotrexate/pharmacokinetics , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyridones/pharmacokinetics , Adult , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/adverse effects , Drug Interactions , Humans , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Middle Aged , Piperazines/administration & dosage , Piperazines/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Pyridones/administration & dosage , Pyridones/adverse effects
13.
Mol Pharm ; 15(4): 1607-1617, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29522347

ABSTRACT

In this study, a multipronged approach of in vitro experiments, in silico simulations, and in vivo studies was developed to evaluate the dissolution, supersaturation, precipitation, and absorption of three formulations of Compound-A, a BCS class 2 weak base with pH-dependent solubility. In in vitro 2-stage dissolution experiments, the solutions were highly supersaturated with no precipitation at the low dose but increasing precipitation at higher doses. No difference in precipitation was observed between the capsules and tablets. The in vitro precipitate was found to be noncrystalline with higher solubility than the crystalline API, and was readily soluble when the drug concentration was lowered by dilution. A gastric transit and biphasic dissolution (GTBD) model was developed to better mimic gastric transfer and intestinal absorption. Precipitation was also observed in GTBD, but the precipitate redissolved and partitioned into the organic phase. In vivo data from the phase 1 clinical trial showed linear and dose proportional PK for the formulations with no evidence of in vivo precipitation. While the in vitro precipitation observed in the 2-stage dissolution appeared to overestimate in vivo precipitation, the GTBD model provided absorption profiles consistent with in vivo data. In silico simulation of plasma concentrations by GastroPlus using biorelevant in vitro dissolution data from the tablets and capsules and assuming negligible precipitation was in line with the observed in vivo profiles of the two formulations. The totality of data generated with Compound-A indicated that the bioavailability differences among the three formulations were better explained by the differences in gastric dissolution than intestinal precipitation. The lack of intestinal precipitation was consistent with several other BCS class 2 basic compounds in the literature for which highly supersaturated concentrations and rapid absorption were also observed.


Subject(s)
Intestinal Absorption/physiology , Pharmaceutical Preparations/metabolism , Tablets/metabolism , Biological Availability , Biopharmaceutics/methods , Chemistry, Pharmaceutical/methods , Computer Simulation , Humans , Intestines/chemistry , Solubility , Stomach/physiology
14.
Clin Pharmacol Ther ; 103(6): 1020-1028, 2018 06.
Article in English | MEDLINE | ID: mdl-29484638

ABSTRACT

GDC-0853 is a small molecule inhibitor of Bruton's tyrosine kinase (BTK) that is highly selective and noncovalent, leading to reversible binding. In double-blind, randomized, and placebo-controlled phase I healthy volunteer studies, GDC-0853 was well tolerated, with no dose-limiting adverse events (AEs) or serious AEs. The maximum tolerated dose was not reached during dose escalation (≤600 mg, single ascending dose (SAD) study; ≤250 mg twice daily (b.i.d.) and ≤500 mg once daily, 14-day multiple ascending dose (MAD) study). Plasma concentrations peaked 1-3 hours after oral administration and declined thereafter, with a steady-state half-life ranging from 4.2-9.9 hours. Independent assays demonstrated dose-dependent BTK target engagement. Based on pharmacokinetic/pharmacodynamic (PK/PD) simulations, a once-daily dosing regimen (e.g., 100 mg, q.d.) is expected to maintain a high level of BTK inhibition over the dosing interval. Taken together, the safety and PK/PD data support GDC-0853 evaluation in rheumatoid arthritis, lupus, and other autoimmune or inflammatory indications.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Adolescent , Adult , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Metabolic Clearance Rate , Middle Aged , Piperazines/administration & dosage , Piperazines/adverse effects , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyridones/administration & dosage , Pyridones/adverse effects , Pyridones/pharmacokinetics , Young Adult
15.
Mol Pharm ; 14(10): 3577-3587, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28834434

ABSTRACT

In this study, two dissolution models were developed to achieve in vitro-in vivo relationship for immediate release formulations of Compound-A, a poorly soluble weak base with pH-dependent solubility and low bioavailability in hypochlorhydric and achlorhydric patients. The dissolution models were designed to approximate the hypo-/achlorhydric and normal fasted stomach conditions after a glass of water was ingested with the drug. The dissolution data from the two models were predictive of the relative in vivo bioavailability of various formulations under the same gastric condition, hypo-/achlorhydric or normal. Furthermore, the dissolution data were able to estimate the relative performance under hypo-/achlorhydric and normal fasted conditions for the same formulation. Together, these biorelevant dissolution models facilitated formulation development for Compound-A by identifying the right type and amount of key excipient to enhance bioavailability and mitigate the negative effect of hypo-/achlorhydria due to drug-drug interaction with acid-reducing agents. The dissolution models use readily available USP apparatus 2, and their broader utility can be evaluated on other BCS 2B compounds with reduced bioavailability caused by hypo-/achlorhydria.


Subject(s)
Achlorhydria/complications , Drug Liberation , Models, Chemical , Administration, Oral , Biological Availability , Chemistry, Pharmaceutical , Drug Interactions , Humans , Hydrogen-Ion Concentration , Solubility , Tablets
16.
Pharmacogenet Genomics ; 23(11): 591-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23982262

ABSTRACT

OBJECTIVE: Nevirapine is an important component of highly active antiretroviral therapy used in the treatment of HIV infection. There is a considerable variation in the pharmacokinetics of nevirapine and this variation can impact the efficacy and toxicity of nevirapine. Although some of this variation can be attributed to environmental factors, the degree to which heritability influences nevirapine pharmacokinetics is unknown. This study aims to estimate how much variation in nevirapine pharmacokinetics is due to genetic factors and to investigate the contribution of selected polymorphisms to this variability. METHODS: Two doses of immediate-release nevirapine were administered to European (n=11) and African American (n=6) participants recruited from the Research in Access to Care in the Homeless cohort. A repeated drug administration method was then used to determine the relative genetic contribution (r(GC)) to variability in nevirapine AUC(0-6 h). Nevirapine plasma levels were quantified using LC/MS/MS. Patients were also genotyped for selected polymorphisms in candidate genes that may influence nevirapine pharmacokinetics. RESULTS: A significant r(GC) for nevirapine AUC(0-6 h) was found in Europeans (P=0.02) and African Americans (P=0.01). A trend toward higher nevirapine AUC(0-6 h) for the CYP2B6 516TT (rs3745274; Q172H) genotype was observed in European Americans (P=0.19). CONCLUSION: This study demonstrates that there is a significant genetic component to variability in nevirapine pharmacokinetics. Although genetic variants such as CYP2B6 polymorphisms attributed to some of this variation, these data suggest that there may be additional genetic factors that influence nevirapine pharmacokinetics.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , HIV Infections/drug therapy , Nevirapine/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adult , Black or African American/genetics , Aged , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/blood , Anti-HIV Agents/therapeutic use , Cytochrome P-450 CYP2B6 , Female , Genetic Variation , Genotype , HIV Infections/genetics , HIV Infections/metabolism , Humans , Male , Middle Aged , Nevirapine/administration & dosage , Nevirapine/blood , Nevirapine/therapeutic use , Pharmacogenetics , Polymorphism, Genetic , White People/genetics
18.
J Infect Dis ; 203(10): 1491-502, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21502085

ABSTRACT

BACKGROUND: Host genetic variation influences human immunodeficiency virus (HIV) infection and progression to AIDS. Here we used clinically well-characterized subjects from 5 pretreatment HIV/AIDS cohorts for a genome-wide association study to identify gene associations with rate of AIDS progression. METHODS: European American HIV seroconverters (n = 755) were interrogated for single-nucleotide polymorphisms (SNPs) (n = 700,022) associated with progression to AIDS 1987 (Cox proportional hazards regression analysis, co-dominant model). RESULTS: Association with slower progression was observed for SNPs in the gene PARD3B. One of these, rs11884476, reached genome-wide significance (relative hazard = 0.3; P =3. 370 × 10(-9)) after statistical correction for 700,022 SNPs and contributes 4.52% of the overall variance in AIDS progression in this study. Nine of the top-ranked SNPs define a PARD3B haplotype that also displays significant association with progression to AIDS (hazard ratio, 0.3; P = 3.220 × 10(-8)). One of these SNPs, rs10185378, is a predicted exonic splicing enhancer; significant alteration in the expression profile of PARD3B splicing transcripts was observed in B cell lines with alternate rs10185378 genotypes. This SNP was typed in European cohorts of rapid progressors and was found to be protective for AIDS 1993 definition (odds ratio, 0.43, P = .025). CONCLUSIONS: These observations suggest a potential unsuspected pathway of host genetic influence on the dynamics of AIDS progression.


Subject(s)
Acquired Immunodeficiency Syndrome/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Regulation/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Polymorphism, Single Nucleotide , Acquired Immunodeficiency Syndrome/pathology , Chromosome Mapping , Disease Progression , Genome, Human , Humans
20.
J Infect Dis ; 202(12): 1836-45, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21083371

ABSTRACT

BACKGROUND: High-throughput genome-wide techniques have facilitated the identification of previously unknown host proteins involved in cellular human immunodeficiency virus (HIV) infection. Recently, 3 independent studies have used small interfering RNA technology to silence each gene in the human genome to determine the importance of each in HIV infection. Genes conferring a significant effect were termed HIV-dependency factors (HDFs). METHODS: We assembled high-density panels of 6380 single-nucleotide polymorphisms (SNPs) in 278 HDF genes and tested for genotype associations with HIV infection and AIDS progression in 1633 individuals from clinical AIDS cohorts. RESULTS: After statistical correction for multiple tests, significant associations with HIV acquisition were found for SNPs in 2 genes, NCOR2 and IDH1. Weaker associations with AIDS progression were revealed for SNPs within the TM9SF2 and EGFR genes. CONCLUSIONS: This study independently verifies the influence of NCOR2 and IDH1 on HIV transmission, and its findings suggest that variation in these genes affects susceptibility to HIV infection in exposed individuals.


Subject(s)
Disease Susceptibility , HIV Infections/genetics , HIV Infections/transmission , HIV-1/pathogenicity , Host-Pathogen Interactions , Isocitrate Dehydrogenase/genetics , Nuclear Receptor Co-Repressor 2/genetics , Disease Progression , ErbB Receptors/genetics , Gene Frequency , Humans , Male , Membrane Proteins/genetics , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...