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










Publication year range
1.
Drug Metab Dispos ; 52(3): 153-158, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38216306

ABSTRACT

The administration of radiolabeled drug candidates is considered the gold standard in absorption, distribution, metabolism, and excretion studies for small-molecule drugs since it allows facile and accurate quantification of parent drug, metabolites, and total drug-related material independent of the compound structure. The choice of the position of the radiolabel, typically 14C or 3H, is critical to obtain relevant information. Sometimes, a biotransformation reaction may lead to cleavage of a part of the molecule. As a result, only the radiolabeled portion can be followed, and information on the fate of the nonlabeled metabolite may be lost. Synthesis and administration of two or more radiolabeled versions of the parent drug as a mixture or in separate studies may resolve this issue but comes with additional challenges. In this paper, we address the questions that may be considered to help make the right choice whether to use a single or multiple radiolabel approach and discuss the pros and cons of different multiple-labeling strategies that can be taken as well as alternative methods that allow the nonlabeled part of the molecule to be followed. SIGNIFICANCE STATEMENT: Radiolabeled studies are the gold standard in drug metabolism research, but molecules can undergo cleavage with loss of the label. This often results in discussions around potential use of multiple labels, which seem to be occurring with increased frequency since an increasing proportion of the small-molecule drugs are tending towards larger molecular weights. This review provides insight and decision criteria in considering a multiple-label approach as well as pros and cons of different strategies that can be followed.


Subject(s)
Pharmaceutical Preparations , Humans , Pharmaceutical Preparations/metabolism , Metabolic Clearance Rate , Biotransformation
2.
Clin Pharmacol Ther ; 115(5): 931-938, 2024 May.
Article in English | MEDLINE | ID: mdl-38018358

ABSTRACT

A review of the use of microdoses and isotopic microtracers for clinical intravenous pharmacokinetic (i.v. PK) data provision is presented. The extent of application of the varied approaches available and the relative merits of each are highlighted with the aim of assisting practitioners in making informed decisions on the most scientifically appropriate design to adopt for any given new drug in development. It is envisaged that significant efficiencies will be realized as i.v. PK data in humans becomes more routinely available for suitable assets in early development, than has been the case prior to the last decade.


Subject(s)
Decision Making , Pharmacokinetics , Humans , Administration, Intravenous , Models, Biological
3.
Clin Pharmacol Ther ; 113(4): 775-781, 2023 04.
Article in English | MEDLINE | ID: mdl-35733280

ABSTRACT

The human absorption, distribution, metabolism, and excretion (hADME) study is the cornerstone of the clinical pharmacology package for small molecule drugs, providing comprehensive information on the rates and routes of disposition and elimination of drug-related material in humans through the use of 14 C-labeled drug. Significant changes have already been made in the design of the hADME study for many companies, but opportunity exists to continue to re-think both the design and timing of the hADME study in light of the potential offered by newer technologies, that enable flexibility in particular to reducing the magnitude of the radioactive dose used. This paper provides considerations on the variety of current strategies that exist across a number of pharmaceutical companies and on some of the ongoing debates around a potential move to the so called "human first/human only" approach, already adopted by at least one company. The paper also provides a framework for continuing the discussion in the application of further shifts in the paradigm.

4.
Drug Metab Dispos ; 50(11): 1442-1453, 2022 11.
Article in English | MEDLINE | ID: mdl-36153007

ABSTRACT

GSK3640254 is a next-generation maturation inhibitor in development for HIV-1 treatment, with pharmacokinetics (PK) supporting once-daily oral dosing in human. This open-label, nonrandomized, two-period clinical mass balance and excretion study was used to investigate the excretion balance, PK, and metabolism of GSK3640254. Five healthy men received a single intravenous microtracer of 100 µg [14C]GSK3640254 with a concomitant oral nonradiolabeled 200-mg tablet followed by an oral 85-mg dose of [14C]GSK3640254 14 days later. Complementary methods, including intravenous microtracing and accelerator mass spectrometry, allowed characterization of several parameters, including fraction absorbed, fraction escaping gut metabolism, hepatic extraction ratio, and renal clearance. Intravenous PK of GSK3640254 was characterized by low plasma clearance (1.04 l/h), moderate terminal phase half-life (21.7 hours), and low volume of distribution at steady state (28.7 L). Orally dosed GSK3640254 was absorbed (fraction absorbed, 0.26), with a high fraction escaping gut metabolism (0.898) and a low hepatic extraction ratio (0.00544), all consistent with low in vitro intrinsic clearance in liver microsomes and hepatocytes. No major metabolites in human plasma required further qualification in animal studies. Both unchanged parent GSK3640254 and its oxidative and conjugative metabolites were excreted into bile, with GSK3640254 likely subject to further metabolism through enterohepatic recirculation. Renal elimination of GSK3640254 as the parent drug or its metabolites was negligible, with >94% of total recovery of oral dose and >99% of the recovered radioactivity in feces. Altogether, the data suggest that systemically available GSK3640254 was slowly eliminated almost entirely by hepatobiliary secretion, primarily as conjugative and oxidative metabolites. SIGNIFICANCE STATEMENT: The combination of an intravenous 14C microtracer with duodenal bile sampling using EnteroTracker in a human absorption, distribution, metabolism, and excretion study enabled derivation of absorption and first-pass parameters, including fraction absorbed, proportion escaping first-pass extraction through the gut wall and liver, hepatic extraction, and other conventional clinical pharmacokinetic parameters. This approach identified hepatic metabolism and biliary excretion as a major elimination pathway for absorbed drug, which would be overlooked based solely on analyses of plasma, urine, and fecal matrices.


Subject(s)
HIV-1 , Administration, Intravenous , Administration, Oral , Animals , Biological Availability , Feces/chemistry , Humans , Male , Metabolic Clearance Rate
5.
Drug Metab Dispos ; 49(12): 1109-1117, 2021 12.
Article in English | MEDLINE | ID: mdl-34625435

ABSTRACT

Linerixibat, an oral small-molecule ileal bile acid transporter inhibitor under development for cholestatic pruritus in primary biliary cholangitis, was designed for minimal absorption from the intestine (site of pharmacological action). This study characterized the pharmacokinetics, absorption, metabolism, and excretion of [14C]-linerixibat in humans after an intravenous microtracer concomitant with unlabeled oral tablets and [14C]-linerixibat oral solution. Linerixibat exhibited absorption-limited flip-flop kinetics: longer oral versus intravenous half-life (6-7 hours vs. 0.8 hours). The short intravenous half-life was consistent with high systemic clearance (61.9 l/h) and low volume of distribution (16.3 l). In vitro studies predicted rapid hepatic clearance via cytochrome P450 3A4 metabolism, which predicted human hepatic clearance within 1.5-fold. However, linerixibat was minimally metabolized in humans after intravenous administration: ∼80% elimination via biliary/fecal excretion (>90%-97% as unchanged parent) and ∼20% renal elimination by glomerular filtration (>97% as unchanged parent). Absolute oral bioavailability of linerixibat was exceedingly low (0.05%), primarily because of a very low fraction absorbed (0.167%; fraction escaping first-pass gut metabolism (fg) ∼100%), with high hepatic extraction ratio (77.0%) acting as a secondary barrier to systemic exposure. Oral linerixibat was almost entirely excreted (>99% recovered radioactivity) in feces as unchanged and unabsorbed linerixibat. Consistent with the low oral fraction absorbed and ∼20% renal recovery of intravenous [14C]-linerixibat, urinary elimination of orally administered radioactivity was negligible (<0.04% of dose). Linerixibat unequivocally exhibited minimal gastrointestinal absorption and oral systemic exposure. Linerixibat represents a unique example of high CYP3A4 clearance in vitro but nearly complete excretion as unchanged parent drug via the biliary/fecal route. SIGNIFICANCE STATEMENT: This study conclusively established minimal absorption and systemic exposure to orally administered linerixibat in humans. The small amount of linerixibat absorbed was eliminated efficiently as unchanged parent drug via the biliary/fecal route. The hepatic clearance mechanism was mispredicted to be mediated via cytochrome P450 3A4 metabolism in vitro rather than biliary excretion of unchanged linerixibat in vivo.


Subject(s)
Administration, Intravenous , Administration, Oral , Carrier Proteins/antagonists & inhibitors , Hepatobiliary Elimination , Membrane Glycoproteins/antagonists & inhibitors , Methylamines/pharmacokinetics , Renal Elimination , Thiazepines/pharmacokinetics , Adult , Biological Availability , Gastrointestinal Agents/pharmacokinetics , Healthy Volunteers , Hepatobiliary Elimination/drug effects , Hepatobiliary Elimination/physiology , Humans , Intestinal Absorption , Male , Metabolic Clearance Rate , Renal Elimination/drug effects , Renal Elimination/physiology , Treatment Outcome
6.
Article in English | MEDLINE | ID: mdl-32071044

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is an inflammatory lung condition, causing progressive decline in lung function leading to premature death. Acute exacerbations in COPD patients are predominantly associated with respiratory viruses. Ribavirin is a generic broad-spectrum antiviral agent that could be used for treatment of viral respiratory infections in COPD. Using the Particle Replication In Nonwetting Templates (PRINT) technology, which produces dry-powder particles of uniform shape and size, two new inhaled formulations of ribavirin (ribavirin-PRINT-CFI and ribavirin-PRINT-IP) were developed for efficient delivery to the lung and to minimize bystander exposure. Ribavirin-PRINT-CFI was well tolerated in healthy participants after single dosing and ribavirin-PRINT-IP was well tolerated in healthy and COPD participants after single and repeat dosing. Ribavirin-PRINT-CFI was replaced with ribavirin-PRINT-IP since the latter formulation was found to have improved physicochemical properties and it had a higher ratio of active drug to excipient per unit dose. Ribavirin concentrations were measured in lung epithelial lining fluid in both healthy and COPD participants and achieved target concentrations. Both formulations were rapidly absorbed with approximately dose proportional pharmacokinetics in plasma. Exposure to bystanders was negligible based on both the plasma and airborne ribavirin concentrations with the ribavirin-PRINT-IP formulation. Thus, ribavirin-PRINT-IP allowed for an efficient and convenient delivery of ribavirin to the lungs while minimizing systemic exposure. Further clinical investigations would be required to demonstrate ribavirin-PRINT-IP antiviral characteristics and impact on COPD viral-induced exacerbations. (The clinical trials discussed in this study have been registered at ClinicalTrials.gov under identifiers NCT03243760 and NCT03235726.).


Subject(s)
Antiviral Agents/administration & dosage , Dry Powder Inhalers , Lung/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Ribavirin/administration & dosage , Administration, Inhalation , Adult , Aged , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Double-Blind Method , Drug Delivery Systems , Dry Powder Inhalers/adverse effects , Female , Healthy Volunteers , Humans , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/virology , Respiratory Mucosa/metabolism , Ribavirin/pharmacokinetics , Ribavirin/therapeutic use , Young Adult
7.
Altern Lab Anim ; 46(6): 335-346, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30657329

ABSTRACT

Phase 0 approaches, including microdosing, involve the use of sub-therapeutic exposures to the tested drugs, thus enabling safer, more-relevant, quicker and cheaper first-in-human (FIH) testing. These approaches also have considerable potential to limit the use of animals in human drug development. Recent years have witnessed progress in applications, methodology, operations, and drug development culture. Advances in applications saw an expansion in therapeutic areas, developmental scenarios and scientific objectives, in, for example, protein drug development and paediatric drug development. In the operational area, the increased sensitivity of Liquid Chromatography Tandem Mass Spectrometry (LC-MS/MS), expansion of the utility of Positron Emission Tomography (PET) imaging, and the introduction of Cavity Ring-Down Spectroscopy (CRDS), have led to the increased accessibility and utility of Phase 0 approaches, while reducing costs and exposure to radioactivity. PET has extended the application of microdosing, from its use as a predominant tool to record pharmacokinetics, to a method for recording target expression and target engagement, as well as cellular and tissue responses. Advances in methodology include adaptive Phase 0/Phase 1 designs, cassette and cocktail microdosing, and Intra-Target Microdosing (ITM), as well as novel modelling opportunities and simulations. Importantly, these methodologies increase the predictive power of extrapolation from microdose to therapeutic level exposures. However, possibly the most challenging domain in which progress has been made, is the culture of drug development. One of the main potential values of Phase 0 approaches is the opportunity to terminate development early, thus not only applying the principle of 'kill-early-kill-cheap' to enhance the efficiency of drug development, but also obviating the need for the full package of animal testing required for therapeutic level Phase 1 studies. Finally, we list developmental scenarios that utilised Phase 0 approaches in novel drug development.


Subject(s)
Animal Experimentation/ethics , Animal Testing Alternatives/ethics , Drug Development/ethics , Drug Development/legislation & jurisprudence , Animal Experimentation/legislation & jurisprudence , Animal Testing Alternatives/legislation & jurisprudence , Animal Welfare/ethics , Animal Welfare/legislation & jurisprudence , Animals , Chromatography, Liquid , Humans , Positron-Emission Tomography , Tandem Mass Spectrometry
9.
Br J Clin Pharmacol ; 78(6): 1185-200, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25041729

ABSTRACT

Human radiolabel studies are traditionally conducted to provide a definitive understanding of the human absorption, distribution, metabolism and excretion (ADME) properties of a drug. However, advances in technology over the past decade have allowed alternative methods to be employed to obtain both clinical ADME and pharmacokinetic (PK) information. These include microdose and microtracer approaches using accelerator mass spectrometry, and the identification and quantification of metabolites in samples from classical human PK studies using technologies suitable for non-radiolabelled drug molecules, namely liquid chromatography-mass spectrometry and nuclear magnetic resonance spectroscopy. These recently developed approaches are described here together with relevant examples primarily from experiences gained in support of drug development projects at GlaxoSmithKline. The advantages of these study designs together with their limitations are described. We also discuss special considerations which should be made for a successful outcome to these new approaches and also to the more traditional human radiolabel study in order to maximize knowledge around the human ADME properties of drug molecules.


Subject(s)
Pharmacokinetics , Carbon Radioisotopes , Chromatography, Liquid , Humans , Magnetic Resonance Spectroscopy , Tandem Mass Spectrometry , Tissue Distribution
10.
Br J Clin Pharmacol ; 78(3): 524-32, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24606567

ABSTRACT

AIMS: The aim of this phase 1, single centre, open label study in four patients with solid tumours was to determine the absolute bioavailability of a 2 mg oral dose of trametinib. Trametinib is an orally bioavailable, reversible and selective allosteric inhibitor of MEK1 and MEK2 activation and kinase activity. METHODS: A microtracer study approach, in which a 5 µg radiolabelled i.v. microdose of trametinib was given concomitantly with an unlabelled 2 mg oral tablet formulation, was used to recover i.v. and oral pharmacokinetic parameters, simultaneously. RESULTS: The least-squares mean (90% confidence interval) absolute bioavailability of trametinib (2 mg tablet) was 72.3% (50.0%, 104.6%). Median tmax after oral administration was 1.5 h and the geometric mean terminal half-life was 11 days. The geometric mean clearance and volume of distribution after i.v. administration were 3.21 l h(-1) and 976 l, respectively, resulting in a terminal elimination half-life of 11 days. CONCLUSIONS: Trametinib absolute bioavailability was moderate to high, whereas first pass metabolism was low.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Administration, Intravenous , Administration, Oral , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biological Availability , Female , Half-Life , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Male , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Pyrimidinones/pharmacokinetics , Pyrimidinones/therapeutic use , Tissue Distribution
11.
AAPS J ; 16(2): 357-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24500278

ABSTRACT

The technique of accelerator mass spectrometry (AMS) is applicable to the analysis of a wide range of trace elemental isotopes. However, in the context of the pharmaceutical industry, it is invariably used to measure radiocarbon ((14)C). There are two broad modes of application: analysis of total (14)C sometimes termed "direct AMS" and analysis of specific (14)C-labelled analytes in a variety of matrices following some method of isolation. It is the latter application which is within the remit of the GBC team, and the team has made efforts to propose harmonized recommendations for the validation of AMS when used in a regulatory bioanalytical mode, i.e. the quantification of specific analyte(s) using liquid chromatography with off-line detection by AMS now known as "LC + AMS". The GBC team has reached a position where they have agreed to many aspects, but also differ on some aspects of what constitutes a bioanalytical assay validation in support of clinical studies using this technology. The detail of most of this will be covered under separate publication(s), but for the purposes of this paper, we have outlined the points of consensus. The purpose of this article is not to provide a roadmap for validation of LC + AMS assays, but to highlight agreements amongst the industry representative experts and the practitioners, as well as identifying specific areas essential for establishing assay quality but where additional discussion is required to reach agreement.


Subject(s)
Biological Assay/methods , Chromatography, Liquid/methods , International Cooperation , Mass Spectrometry/methods
12.
Drug Metab Dispos ; 42(3): 415-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24378325

ABSTRACT

The absorption, metabolism, and excretion of darapladib, a novel inhibitor of lipoprotein-associated phospholipase A2, was investigated in healthy male subjects using [(14)C]-radiolabeled material in a bespoke study design. Disposition of darapladib was compared following single i.v. and both single and repeated oral administrations. The anticipated presence of low circulating concentrations of drug-related material required the use of accelerator mass spectrometry as a sensitive radiodetector. Blood, urine, and feces were collected up to 21 days post radioactive dose, and analyzed for drug-related material. The principal circulating drug-related component was unchanged darapladib. No notable metabolites were observed in plasma post-i.v. dosing; however, metabolites resulting from hydroxylation (M3) and N-deethylation (M4) were observed (at 4%-6% of plasma radioactivity) following oral dosing, indicative of some first-pass metabolism. In addition, an acid-catalyzed degradant (M10) resulting from presystemic hydrolysis was also detected in plasma at similar levels of ∼5% of radioactivity post oral dosing. Systemic exposure to radioactive material was reduced within the repeat dose regimen, consistent with the notion of time-dependent pharmacokinetics resulting from enhanced clearance or reduced absorption. Elimination of drug-related material occurred predominantly via the feces, with unchanged darapladib representing 43%-53% of the radioactive dose, and metabolites M3 and M4 also notably accounting for ∼9% and 19% of the dose, respectively. The enhanced study design has provided an increased understanding of the absorption, distribution, metabolism and excretion (ADME) properties of darapladib in humans, and substantially influenced future work on the compound.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , Benzaldehydes/metabolism , Oximes/metabolism , Phospholipase A2 Inhibitors/metabolism , Administration, Oral , Adult , Benzaldehydes/administration & dosage , Benzaldehydes/blood , Benzaldehydes/pharmacokinetics , Biotransformation , Carbon Isotopes , Carbon Radioisotopes , Feces/chemistry , Humans , Injections, Intravenous , Male , Metabolic Clearance Rate , Molecular Structure , Oximes/administration & dosage , Oximes/blood , Oximes/pharmacokinetics , Phospholipase A2 Inhibitors/administration & dosage , Phospholipase A2 Inhibitors/blood , Phospholipase A2 Inhibitors/pharmacokinetics , Tissue Distribution
13.
J Clin Pharmacol ; 53(9): 955-61, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23846776

ABSTRACT

Dabrafenib is an orally bioavailable, potent, and selective inhibitor of human wild-type BRAF and CRAF kinases as well as mutant forms of BRAF kinase. The aim of this phase 1, single-center, open-label study in four patients with BRAF mutation-positive solid tumors was to determine the absolute bioavailability of a 150 mg oral dose of dabrafenib. A microtracer study approach, in which a 50 µg radiolabeled intravenous (IV) microdose of dabrafenib was given concomitantly with a 150 mg oral dose, was used to simultaneously recover IV and oral pharmacokinetic parameters. The least squares mean (90% CI) absolute bioavailability of dabrafenib (HPMC capsules) was 94.5% (81.3%, 109.7%). Median T(max) after oral administration was 2.0 hours and the geometric mean terminal half-life was 4.8 hours. The geometric mean clearance and volume of distribution after IV administration were 12.0 L/h and 45.5 L, respectively. Human clearance and volume of distribution at steady state were in agreement with predictions made using allometric scaling of pharmacokinetic parameters from four preclinical species. In conclusion, dabrafenib absolute bioavailability was high, whereas first-pass metabolism was low. Furthermore, the microtracer approach provided an innovative and efficient method for assessing the absolute bioavailability of dabrafenib in patients with advanced cancer.


Subject(s)
Imidazoles/pharmacokinetics , Neoplasms/metabolism , Oximes/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Administration, Intravenous , Administration, Oral , Biological Availability , Female , Humans , Imidazoles/administration & dosage , Imidazoles/blood , Male , Mutation , Oximes/administration & dosage , Oximes/blood , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Proto-Oncogene Proteins B-raf/genetics
14.
Drug Metab Dispos ; 41(5): 1070-81, 2013 May.
Article in English | MEDLINE | ID: mdl-23439661

ABSTRACT

(S)-3-(Aminomethyl)-7-(3-hydroxypropoxy)-1-hydroxy-1,3-dihydro-2,1-benzoxaborole (GSK2251052) is a novel boron-containing antibiotic that inhibits bacterial leucyl tRNA synthetase, and that has been in development for the treatment of serious Gram-negative infections. In this study, six healthy adult male subjects received a single i.v. dose of [¹4C]GSK2251052, 1500 mg infused over 1 hour. Blood, urine, and feces were collected over an extended period of 14 days, and accelerator mass spectrometry was used to quantify low levels of radioactivity in plasma at later time points to supplement the less-sensitive liquid scintillation counting technique. An excellent mass balance recovery was achieved representing a mean total of 98.2% of the dose, including 90.5% recovered in the urine. Pharmacokinetic analysis demonstrated that radioactivity was moderately associated with the blood cellular components, and together with GSK2251052, both were highly distributed into tissues. The parent compound had a much shorter half-life than total radioactivity in plasma, approximately 11.6 hours compared with 96 hours. GSK2251052 and its major metabolite M3, which resulted from oxidation of the propanol side chain to the corresponding carboxylic acid, comprised the majority of the plasma radioactivity, 37 and 53% of the area under the plasma versus time concentration curve from time zero to infinity, respectively. Additionally, M3 was eliminated renally, and was demonstrated to be responsible for the long plasma radioactivity elimination half-life. A combination of in vitro metabolism experiments and a pharmacokinetic study in monkeys with the inhibitor 4-methylpyrazole provided strong evidence that alcohol dehydrogenase, potentially in association with aldehyde dehydrogenase, is the primary enzyme involved in the formation of the M3 metabolite.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Boron Compounds/pharmacokinetics , Boron/analysis , Animals , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/urine , Boron Compounds/blood , Boron Compounds/urine , Humans , Macaca fascicularis , Male , Mass Spectrometry
15.
Drug Metab Dispos ; 41(1): 89-100, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23043183

ABSTRACT

The metabolism and disposition of vilanterol, a novel long-acting ß(2)-adrenoceptor agonist (LABA) for inhalation use, was investigated after oral administration in humans. Single oral administrations of up to 500 µg of vilanterol were shown to be safe and well tolerated in two clinical studies in healthy men. In a human radiolabel study, six healthy men received a single oral dose of 200 µg of [(14)C]vilanterol (74 kBq). Plasma, urine, and feces were collected up to 168 hours after the dose and were analyzed for vilanterol, metabolites, and radioactivity. At least 50% of the radioactive dose was orally absorbed. The primary route of excretion of drug-related material was via O-dealkylation to metabolites, which were mainly excreted in urine. Vilanterol represented a very small percentage (<0.5%) of the total drug-related material in plasma, indicative of extensive first-pass metabolism. Circulating metabolites resulted mainly from O-dealkylation and exhibited negligible pharmacologic activity. The therapeutic dose level for vilanterol is 25 µg by the inhalation route. At this low-dose level, the likelihood of pharmacologically inactive metabolites causing unexpected toxicity is negligible. In addition to providing an assessment of the disposition of vilanterol in human, this work highlights a number of complexities associated with determining human absorption, distribution, metabolism, and excretion (ADME) for inhaled molecules--mainly related to the low chemical doses and complications associated with the inhalation route of administration.


Subject(s)
Adrenergic beta-Antagonists/pharmacokinetics , Benzyl Alcohols/pharmacokinetics , Chlorobenzenes/pharmacokinetics , Administration, Inhalation , Adrenergic beta-Antagonists/administration & dosage , Animals , Benzyl Alcohols/administration & dosage , Carbon Radioisotopes , Chlorobenzenes/administration & dosage , Chromatography, High Pressure Liquid , Dogs , Humans , Male , Mass Spectrometry , Mice , Rabbits , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...