Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Arthritis Res Ther ; 24(1): 199, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35982464

ABSTRACT

BACKGROUND: Mitogen-activated protein kinase (MAPK)-activated protein kinase-2 (MK2) is activated downstream of p38 MAPK and regulates stability of mRNAs encoding inflammatory cytokines. CC-99677 is a novel, irreversible, covalent MK2 inhibitor under development for the treatment of ankylosing spondylitis (AS) and other inflammatory diseases. As part of a phase I clinical trial to assess safety and tolerability, we evaluated target engagement, pharmacokinetics, and pharmacodynamics of CC-99677. METHODS: The MK2 inhibitor CC-99677 was evaluated for its effect on cytokine expression in vitro in peripheral blood mononuclear cells (PBMCs) from healthy donors and patients with a definitive AS diagnosis. A novel in vitro model was developed to compare the potential for tachyphylaxis of CC-99677 and p38 inhibitors in THP-1 cells. The effect of CC-99677 on tristetraprolin (TTP) and cytokine mRNA was assessed in stimulated human monocyte-derived macrophages. In a first-in-human study, thirty-seven healthy volunteers were randomly assigned to daily oral doses of CC-99677 or placebo, and blood was collected at pre-specified time points before and after dosing. CC-99677 concentrations were assessed in the plasma, and CC-99677 binding to MK2 was evaluated in PBMCs. Ex vivo stimulation of the whole blood was conducted from participants in the first-in-human study to assess the pharmacodynamic effects. RESULTS: In vitro, CC-99677 inhibited tumor necrosis factor (TNF), interleukin (IL)-6, and IL-17 protein production in samples of monocytes and macrophages from AS patients and healthy volunteers via an mRNA-destabilization mechanism. In the in vitro model of tachyphylaxis, CC-99677 showed a differentiated pattern of sustained TNF protein inhibition compared with p38 inhibitors. CC-99677 reduced TTP phosphorylation and accelerated the decay of inflammatory cytokine mRNA in lipopolysaccharide-stimulated macrophages. Administration of CC-99677 to healthy volunteers was safe and well-tolerated, with linear pharmacokinetics and sustained reduction of ex vivo whole blood TNF, IL-6, and chemokine synthesis. CONCLUSIONS: CC-99677 inhibition of MK2 is a promising approach for the treatment of inflammatory diseases and may overcome the limitations of p38 MAPK inhibition. TRIAL REGISTRATION: ClinicalTrials.gov NCT03554993 .


Subject(s)
Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Cytokines/metabolism , Humans , Leukocytes, Mononuclear/metabolism , RNA, Messenger , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Front Pharmacol ; 11: 580560, 2020.
Article in English | MEDLINE | ID: mdl-33364955

ABSTRACT

The Association for Human Pharmacology in the Pharmaceutical Industry (AHPPI) annual meeting focused on impending change, innovation, and future challenges facing early phase drug development as we move into the second decade of the 21th century. The meeting opened with discussion around the technical revolution in pharmaceutical medicine over the 4 decades since the AHPPI was founded and how transformative technologies have accompanied the introduction of processes such as physiologically based pharmacokinetic modeling. During the meeting examples were presented of how in terms of the development of new therapies, the classic phases of clinical drug development are becoming a thing of the past and the lines between the phases have begun to blur, particularly in the field of oncology. The contribution that monoclonal antibodies have made to medicine and the next chapter in their design and use was also discussed. A representative of the UK's Medicine and Healthcare Products Regulatory Agency discussed the increasing numbers of requests to approve complex innovative design trials, how novel trial designs are impacting on the traditional linear "phase" approach to drug development and the common pitfalls associated with them. Guidance was provided from a regulator's viewpoint on what was meant by the term "novel design" and how to submit successful trial applications for such complex trials. In an Oxford-style debate, the audience discussed the motion that "there is no longer a need to include placebo subjects in early clinical trials." The keynote speaker focused on delivering change in complex environments such as the field of drug development. The afternoon session included presentations on the challenges associated with drug product design, the complexities within non-oral dosage forms and proposed new methods of formulations for drug delivery. Presentations were also given on advances in mechanistic and computational pharmacokinetic modeling and how they have proved to be valuable tools to rationalize and facilitate the process of drug development.

3.
Neurogastroenterol Motil ; 32(3): e13784, 2020 03.
Article in English | MEDLINE | ID: mdl-32017341

ABSTRACT

BACKGROUND: Ulimorelin, a small molecule ghrelin agonist and prokinetic agent, was effective in animal models of gastroparesis and delayed transit. However, employing once daily administration, it failed in clinical trials of postoperative ileus (POI), a condition in which colonic motility recovers last. The aim of this study was to evaluate drug dosing and regional differences in drug activity between stomach and colon. METHODS: Gastric emptying was assessed by scintigraphy in healthy adults at single doses of 600-1200 µg kg-1 and multiple doses of 80-600 µg kg-1 Q8H for 7 days. Colonic motility was assessed by 7-region scintigraphic analysis at a dose of 600 µg kg-1 for 2 days. The primary endpoints were percent change in time to 50% (∆t50 ) liquid gastric emptying on Days 1, 4, and 6 and the geometric mean center of colonic transit at 24 hours (GC24 ). Plasma concentrations of free and total ulimorelin were measured for pharmacokinetic and exposure-response modeling. KEY RESULTS: Ulimorelin 150-600 µg kg-1 every 8 hours resulted in statistically significant improvements (∆t50  = 23% to 46% (P < .05)) in gastric emptying from baseline that were sustained through Day 6. However, no effects on GC24 were observed. Pharmacokinetic analyses suggested that the free concentrations of ulimorelin achieved in POI trials and dosing frequency may have been inadequate. CONCLUSIONS AND INFERENCES: Ulimorelin is a potent gastric prokinetic but lacks evidence of activity in the human colon, pointing to the stomach as the predominant site of action of ghrelin in humans; ClinicalTrials.gov NCT02993055.


Subject(s)
Gastric Emptying/drug effects , Gastrointestinal Transit/drug effects , Macrocyclic Compounds/pharmacology , Adult , Female , Ghrelin , Humans , Male
4.
J Cachexia Sarcopenia Muscle ; 10(5): 1027-1035, 2019 10.
Article in English | MEDLINE | ID: mdl-31074178

ABSTRACT

BACKGROUND: Loss of appetite and body weight are potentially devastating, highly prevalent cancer complications. The ghrelin receptor is a mediator of appetite and metabolism, and anamorelin is a novel, orally administered ghrelin receptor agonist. Effects on appetite and food intake may influence body-weight gain but can be difficult to measure in multi-site studies. Here, we summarize two single-centre trials. METHODS: Both trials were phase I, randomized, double-blind, placebo-controlled, partly/wholly crossover studies of healthy young adults. Study 102 tested single anamorelin doses of 1-20 mg. Assessments included post-dose self-ratings on 100 mm visual analogue scales for hunger, anticipated eating pleasure, and anticipated quantity of food consumption. Study 101 tested single 10, 25, and 50 mg doses. Assessments included the same scales plus caloric intake beginning 4 h post-dose. RESULTS: Study 102 treated 16 male subjects (mean age, 26.3 years). Mean hunger scores generally increased after all treatments, with significant differences from placebo (P < 0.05) in the 5 mg anamorelin group at 0.5 and 1 h post-dose (+8.2 and +13.2 mm). Results for other scales were similar. Study 101 treated nine male subjects (mean age, 26.3 years). Pooled findings for anamorelin 25 and 50 mg vs. placebo showed significant mean increases in hunger scores at all but 1 pre-prandial time point, including the first assessment, 0.5 h post-dose (+10.9 vs. +0.7 mm, P = 0.0077), and the last assessment, 4 h post-dose (+32.7 vs. +7.0 mm, P = 0.0170), with a significant mean 18.4% increase vs. placebo in caloric intake (P = 0.0148). CONCLUSIONS: In single-centre studies of healthy adults, single anamorelin doses of 1-20 mg elicited modest increases in hunger, and single doses of 25 and 50 mg achieved significant increases in hunger and caloric intake. The findings are consistent with dose-related weight gain reported in a prior phase I study as well as multi-centre findings in cachectic cancer patients and expand the evidence supporting anamorelin as a potential intervention.


Subject(s)
Appetite/drug effects , Eating/drug effects , Hydrazines/therapeutic use , Oligopeptides/therapeutic use , Administration, Oral , Adult , Female , Humans , Hydrazines/administration & dosage , Hydrazines/adverse effects , Male , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Treatment Outcome , Young Adult
5.
AAPS PharmSciTech ; 20(2): 90, 2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30684094

ABSTRACT

Celecoxib (Celebrex®) is the only widely used NSAID that selectively inhibits the COX-2 isoenzyme. Celebrex® is absorbed slowly in the fasted state and food intake further delays absorption. In this work, an amorphous water dispersible granule formulation of celecoxib is described with in vitro characterization, preclinical and clinical data. The formulation exhibited very high passive permeability and apparent solubility, significantly outperforming the micronized celecoxib and the drug product Celebrex®. The granule formulation remained stable for at least 1 year in stability tests. In dog studies, tmax was 1 h with over 50% of Cmax reached within 15 min regardless of food intake. A phase 1 clinical trial was conducted with 12 volunteers at 100- and 200-mg doses. Celecoxib plasma concentrations reached 250 ng/ml, the effective therapeutic plasma level, in less than 15 min regardless of food or dose. The novel celecoxib formulation is rapidly absorbed, demonstrating the potential utility as an acute treatment offering advantages over the currently marketed product.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Celecoxib/chemistry , Adolescent , Adult , Aged , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Celecoxib/pharmacokinetics , Drug Compounding , Drug Stability , Humans , Male , Middle Aged , Permeability , Solubility , Young Adult
6.
Xenobiotica ; 49(9): 1033-1043, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30231665

ABSTRACT

The absorption, metabolism and excretion of MT-1303 were investigated in healthy male subjects after a single oral dose of 0.4 mg [14C]-MT-1303 (ClinicalTrials.gov NCT02293967). The MT-1303 concentration in the plasma reached a maximum at 12 h after administration. Thereafter, the concentration declined with a half-life of 451 h. At the final assessment on Day 57, 91.16% of the administered radioactivity was excreted, and the cumulative excretion in the urine and faeces was 35.32% and 55.84%, respectively. The most abundant metabolite in plasma was MT-1303-P, which accounted for 42.6% of the area under the plasma concentration-time curve (AUC) of the total radioactivity. The major component excreted in urine was Human Urine (HU)4 (3066434), accounting for 28.1% of radioactivity in the sample (4.05% of the dose), whereas MT-1303 was a major component in the faeces, accounting for 89.8% of radioactivity in the sample (25.49% of the dose) up to 240 h after administration. This study indicates that multiple metabolic pathways are involved in the elimination of MT-1303 from the human body and the excretion of MT-1303 and MT-1303-P via the kidney is low. Therefore, MT-1303 is unlikely to cause conspicuous drug interactions or alter pharmacokinetics in patients with renal impairment.


Subject(s)
Propanolamines/pharmacokinetics , Administration, Oral , Carbon Radioisotopes/administration & dosage , Carbon Radioisotopes/pharmacokinetics , Feces , Half-Life , Healthy Volunteers , Humans , Inactivation, Metabolic , Male , Middle Aged , Propanolamines/administration & dosage , Propanolamines/blood , Propanolamines/urine , Tissue Distribution
7.
Front Pharmacol ; 9: 1301, 2018.
Article in English | MEDLINE | ID: mdl-30510508

ABSTRACT

The Association for Human Pharmacology in the Pharmaceutical Industry (AHPPI) annual meeting focused on the changing face of early phase drug development and opened with a keynote speech concerning the revolution in pharmaceutical medicine over the last 30 years and the impact this has had on the way patients are treated. Examples were presented of how translational pharmaceutics is being used to tackle the high drug candidate failure rate and is improving productivity when moving drug candidates from the laboratory through to clinical proof of concept. The European Medicines Agency revised 2007 Risk Mitigation guideline on first in human (FIH) clinical trials was discussed. The focus of the revised guideline, which came into force in February 2018, is on risk mitigation and promotion of safety and will assist drug sponsors with the design and performance of early clinical studies. The use of integrated adaptive protocol designs in early clinical development was discussed in relation to the challenges involved when running early phase clinical trials in patients. The Health Regulatory Authority presented its strategies to ensure that following Brexit, the United Kingdom remains an attractive place to conduct Phase I clinical trials. The Medicines and Healthcare products Regulatory Agency confirmed that in the event of a "no deal" Brexit, it is well placed to implement and influence many provisions of the new EU CTR. The meeting provided an opportunity to discuss the changing regulatory environment and the opportunities and challenges facing the United Kingdom following Brexit with invited speakers from a range of disciplines including drug development, clinical trials and research organizations, government science policy and regulatory agencies.

8.
Cancer Chemother Pharmacol ; 80(4): 723-728, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28776077

ABSTRACT

PURPOSE: Zytiga (abiraterone acetate, AA) is known to exhibit very low bioavailability and a significant positive food effect in men. The unfavorable pharmacokinetic properties are attributed to the inadequate and variable dissolution of the compound. Using a continuous flow precipitation technology, a novel AA formulation has been developed with improved solubility and dissolution characteristics. The current study was performed to evaluate the pharmacokinetics and safety of this novel formulation in healthy volunteers. METHODS: The study was conducted in 11 healthy men aged 47-57 years. All subjects received 3 consecutive single doses of the novel formulation of AA (100 and 200 mg in the fasted state and 200 mg in the fed state). Data were compared with pharmacokinetic and safety data reported for 1000 mg Zytiga, the marketed drug. RESULTS: The novel formulation of AA allows rapid absorption of the compound with t max values within 1 hour. Based on AUC values, a ~250 mg dose of the novel formulation is predicted to give the same exposure as 1000 mg Zytiga in the fasted state. The significant positive food effect was also eliminated; actually, a slight, but statistically significant negative food effect was observed. Variability of exposure was significantly reduced when compared to Zytiga. AA administered in the novel formulation was well tolerated with no IMP-related safety AEs reported. CONCLUSION: The novel formulation might allow a 75% dose reduction with significant reduction of inter-individual variability. The negative food effect observed requires further investigations; however, elimination of the significant positive food effect could be adequate to negate the restriction of a food label.


Subject(s)
Abiraterone Acetate/administration & dosage , Antineoplastic Agents/administration & dosage , Chemistry, Pharmaceutical , Food-Drug Interactions , Abiraterone Acetate/adverse effects , Abiraterone Acetate/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Biological Availability , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Liberation , Humans , Male , Middle Aged , Solubility
9.
Clin Ther ; 38(11): 2447-2458, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27751676

ABSTRACT

PURPOSE: Selumetinib (AZD6244, ARRY-142886), an oral mitogen activated kinase 1/2 inhibitor, is in clinical development for the treatment of a variety of different tumor types. Herein, we report a study that determined the distribution, metabolism, and excretion of selumetinib in healthy male volunteers. METHODS: In this open-label, single-center, Phase I clinical trial, 6 subjects received a single 75-mg dose of [14C]-selumetinib. Blood and excreta samples were collected for pharmacokinetic and radiometric analyses. Tolerability monitoring was performed throughout the study. FINDINGS: The Cmax of plasma selumetinib was 1520 ng/mL at 1 hour postdose and declined with a t1/2 of 13.7 hours. Over a 216-hour postdose collection period, total dose recovery was 93% of the radioactive dose, with 59% recovered from feces and 33% from urine. Circulating drug-related material was primarily associated with plasma, with minimal distribution into red blood cells. Selumetinib was the major circulating drug-related component and accounted for 40% of the plasma radioactivity (mean of AUC0-72h pool). The major circulating metabolite (M2; accounting for 22% of the plasma radioactivity) resulted from multiple biotransformation pathways, including loss of the ethanediol moiety in combination with glucuronidation. A further 6 circulating metabolites were identified, each accounting for between 2% and 7% of plasma radioactivity. Selumetinib was a minor component in urine, accounting for ≤1% of the dose. M2 was the most abundant metabolite in urine, accounting for 10% of the dose, and there were 5 other metabolites accounting for between 1% and 10% of the dose. In feces, selumetinib accounted for a mean of 19% of the dose. Also present were 7 metabolites accounting for between 1% and 9% of the dose. The majority of the dose was recovered as metabolites, indicating that the liver is the major route of drug elimination. There were no tolerability concerns. IMPLICATIONS: The findings from this study will inform the label and will contribute to the understanding of the clinical pharmacology of selumetinib. ClinicalTrials.gov identifier: NCT01931761.


Subject(s)
Benzimidazoles/pharmacokinetics , Administration, Oral , Carbon Radioisotopes , Feces , Humans , Male , Middle Aged
10.
Xenobiotica ; 45(12): 1054-62, 2015.
Article in English | MEDLINE | ID: mdl-25986539

ABSTRACT

1. The pharmacokinetics and disposition of delafloxacin was investigated following a single intravenous (300 mg, 100 µCi) dose to healthy male subjects. 2. Mean Cmax, AUC0-∞, Tmax and t1/2 values for delafloxacin were 8.98 µg/mL, 21.31 µg h/mL, 1 h and 2.35 h, respectively, after intravenous dosing. 3. Radioactivity was predominantly excreted via the kidney with 66% of the radioactive dose recovered in the urine. Approximately 29% of the radioactivity was recovered in the faeces, giving an overall mean recovery of 94% administered radioactivity. 4. The predominant circulating components were identified as delafloxacin and a direct glucuronide conjugate of delafloxacin.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Fluoroquinolones/pharmacokinetics , Administration, Intravenous , Adult , Aged , Anti-Bacterial Agents/administration & dosage , Area Under Curve , Chromatography, High Pressure Liquid , Feces/chemistry , Fluoroquinolones/administration & dosage , Glucuronides/metabolism , Healthy Volunteers , Humans , Kidney/metabolism , Male , Middle Aged , Tissue Distribution
11.
Drug Metab Dispos ; 42(5): 932-42, 2014 May.
Article in English | MEDLINE | ID: mdl-24616266

ABSTRACT

Avibactam, a novel non-ß-lactam ß-lactamase inhibitor with activity against Ambler class A, class C, and some class D enzymes is being evaluated in combination with various ß-lactam antibiotics to treat serious bacterial infections. The in vivo mass balance recovery and metabolite profile of [(14)C] avibactam (500 mg/1-h infusion) was assessed in six healthy male subjects, and a series of in vitro experiments evaluated the metabolism and drug-drug interaction potential of avibactam. In the mass balance study, measurement of plasma avibactam (using a validated liquid chromatography-tandem mass spectrometry method) and total radioactivity in plasma, whole blood, urine, and feces (using liquid scintillation counting) indicated that most of the avibactam was excreted unchanged in urine within 12 hours, with recovery complete (>97% of the administered dose) within 96 hours. Geometric mean avibactam renal clearance (158 ml/min) was greater than the product of unbound fraction of drug and glomerular filtration rate (109.5 ml/min), suggesting that active tubular secretion accounted for some renal elimination. There was no evidence of metabolism in plasma and urine, with unchanged avibactam the major component in both matrices. Avibactam demonstrated in vitro substrate potential for organic anion transporters 1 and 3 (OAT1 and OAT3) proteins expressed in human embryonic kidney 293 cells (Km > 1000 µM; >10-fold the Cmax of a therapeutic dose), which could account for the active tubular secretion observed in vivo. Avibactam uptake by OAT1 and OAT3 was inhibited by probenecid, a potent OAT1/OAT3 inhibitor. Avibactam did not interact with various other membrane transport proteins or cytochrome P450 enzymes in vitro, suggesting it has limited propensity for drug-drug interactions involving cytochrome P450 enzymes.


Subject(s)
Anti-Bacterial Agents/metabolism , Azabicyclo Compounds/metabolism , beta-Lactamase Inhibitors , Adult , Animals , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/urine , Azabicyclo Compounds/blood , Azabicyclo Compounds/urine , Biological Transport , Carbon Radioisotopes , Cell Membrane/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dogs , Drug Interactions , Feces/chemistry , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Metabolic Clearance Rate , Microsomes/drug effects , Microsomes/enzymology , Microsomes/metabolism , Middle Aged , Rabbits , Substrate Specificity
12.
J Clin Pharmacol ; 54(1): 97-108, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24122871

ABSTRACT

Netupitant is a new, selective NK1 receptor antagonist under development for the prevention of chemotherapy-induced nausea and vomiting. Two studies were conducted to evaluate the brain receptor occupancy (RO) and disposition (ADME) of netupitant in humans. Positron emission tomography (PET) imaging with the NK1 receptor-binding-selective tracer [(11) C]-GR205171 was used to evaluate the brain penetration of different doses of netupitant (100, 300, and 450 mg) and to determine the NK1 -RO duration. A NK1 -RO of 90% or higher was achieved with all doses in the majority of the tested brain regions at Cmax, with a long duration of RO. The netupitant minimal plasma concentration predicted to achieve a NK1 -RO of 90%, C90% , in the striatum was 225 ng/mL; after administration of netupitant 300 mg, concentrations exceeded the C90% . In the ADME study, a single nominal dose of [(14) C]-netupitant 300 mg was used to assess its disposition. Absorption was rapid and netupitant was extensively metabolized via Phase I and II hepatic metabolism. Elimination of >90% was predicted at day 29 and was principally via hepatic/biliary route (>85%) with a minor contribution of the renal route (<5%). In conclusion, these studies demonstrate that netupitant is a potent agent targeting NK1 receptors with long lasting RO. In addition, netupitant is extensively metabolized and is mainly eliminated through the hepatic/biliary route and to a lesser extent via the kidneys.


Subject(s)
Biomarkers/analysis , Isoquinolines/pharmacokinetics , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Positron-Emission Tomography/methods , Pyridines/pharmacokinetics , Quinuclidines/pharmacokinetics , Serotonin 5-HT3 Receptor Antagonists/pharmacokinetics , Adult , Area Under Curve , Bile/metabolism , Drug Combinations , Humans , Isoquinolines/administration & dosage , Isoquinolines/pharmacology , Kidney/metabolism , Liver/metabolism , Male , Middle Aged , Neurokinin-1 Receptor Antagonists/administration & dosage , Neurokinin-1 Receptor Antagonists/pharmacology , Palonosetron , Piperidines , Pyridines/administration & dosage , Pyridines/pharmacology , Quinuclidines/administration & dosage , Quinuclidines/pharmacology , Radiopharmaceuticals , Serotonin 5-HT3 Receptor Antagonists/administration & dosage , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Tetrazoles , Tissue Distribution , Young Adult
13.
Xenobiotica ; 44(3): 242-53, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23855261

ABSTRACT

1. The absorption, metabolism and excretion of teneligliptin were investigated in healthy male subjects after a single oral dose of 20 mg [(14)C]teneligliptin. 2. Total plasma radioactivity reached the peak concentration at 1.33 h after administration and thereafter disappeared in a biphasic manner. By 216 h after administration, ≥90% of the administered radioactivity was excreted, and the cumulative excretion in the urine and faeces was 45.4% and 46.5%, respectively. 3. The most abundant metabolite in plasma was a thiazolidine-1-oxide derivative (designated as M1), which accounted for 14.7% of the plasma AUC (area under the plasma concentration versus time curve) of the total radioactivity. The major components excreted in urine were teneligliptin and M1, accounting for 14.8% and 17.7% of the dose, respectively, by 120 h, whereas in faeces, teneligliptin was the major component (26.1% of the dose), followed by M1 (4.0%). 4. CYP3A4 and FMO3 are the major enzymes responsible for the metabolism of teneligliptin in humans. 5. This study indicates the involvement of renal excretion and multiple metabolic pathways in the elimination of teneligliptin from the human body. Teneligliptin is unlikely to cause conspicuous drug interactions or changes in its pharmacokinetics patients with renal or hepatic impairment, due to a balance in the elimination pathways.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Dipeptidyl-Peptidase IV Inhibitors/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Oxygenases/metabolism , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Thiazolidines/metabolism , Thiazolidines/pharmacokinetics , Absorption , Adult , Area Under Curve , Carbon Radioisotopes/blood , Carbon Radioisotopes/pharmacokinetics , Chromatography, High Pressure Liquid , Dipeptidyl-Peptidase IV Inhibitors/blood , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Feces/chemistry , Humans , Male , Mass Spectrometry , Middle Aged , Models, Biological , Molecular Structure , Pyrazoles/blood , Pyrazoles/chemistry , Scintillation Counting , Thiazolidines/blood , Thiazolidines/chemistry , Time Factors
14.
Xenobiotica ; 44(6): 522-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24304170

ABSTRACT

1. Gemigliptin (formerly known as LC15-0444) is a newly developed dipeptidyl peptidase 4 inhibitor for the treatment of type 2 diabetes. Following oral administration of 50 mg (5.4 MBq) [(14)C]gemigliptin to healthy male subjects, absorption, metabolism and excretion were investigated. 2. A total of 90.5% of administered dose was recovered over 192 hr postdose, with 63.4% from urine and 27.1% from feces. Based on urinary recovery of radioactivity, a minimum 63.4% absorption from gastrointestinal tract could be confirmed. 3. Twenty-three metabolites were identified in plasma, urine and feces. In plasma, gemigliptin was the most abundant component accounting for 67.2% ∼ 100% of plasma radioactivity. LC15-0636, a hydroxylated metabolite of gemigliptin, was the only human metabolite with systemic exposure more than 10% of total drug-related exposure. Unchanged gemigliptin accounted for 44.8% ∼ 67.2% of urinary radioactivity and 27.7% ∼ 51.8% of fecal radioactivity. The elimination of gemigliptin was balanced between metabolism and excretion through urine and feces. CYP3A4 was identified as the dominant CYP isozyme converting gemigliptin to LC15-0636 in recombinant CYP/FMO enzymes.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/metabolism , Gastrointestinal Absorption , Piperidones/metabolism , Pyrimidines/metabolism , Administration, Oral , Adult , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Dipeptidyl-Peptidase IV Inhibitors/blood , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Feces/chemistry , Humans , Male , Metabolic Networks and Pathways , Middle Aged , Piperidones/blood , Piperidones/chemistry , Piperidones/pharmacokinetics , Protein Binding , Pyrimidines/blood , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics
15.
Ann Hematol ; 89(10): 971-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20567823

ABSTRACT

Further to the patent expiry of Neupogen (Amgen filgrastim), Hospira has developed a biosimilar filgrastim (Nivestim) that may offer a clinically effective alternative for multiple hematologic and oncologic indications. Here results are reported from a phase I trial, primarily designed to compare the pharmacodynamic profiles of Hospira filgrastim and Amgen filgrastim. A phase I, single-center, double-blind, randomized trial was undertaken to demonstrate equivalence of the pharmacodynamic characteristics of Hospira filgrastim and Amgen filgrastim. Fifty healthy volunteers were randomized to receive 5 or 10 microg/kg dosing, before further randomization to treatment sequence. All volunteers received five daily subcutaneous doses of Hospira filgrastim or Neupogen, with subsequent crossover to the alternative treatment. Bioequivalence was evaluated by analysis of variance; if the estimated 90% confidence intervals (CIs) for the ratio of 'test' to 'reference' treatment means were within the conventional equivalence limits of 0.80-1.25, then bioequivalence was concluded. Forty-eight volunteers completed the study. Geometric mean absolute neutrophil count area under the curve from time 0 to the last time point at day 5 (primary endpoint) was comparable in volunteers given Hospira filgrastim or Amgen filgrastim at 5 microg/kg (ratio of means, 0.98; 90% CI, 0.92-1.05) or 10 microg/kg (ratio, 0.97; 90% CI, 0.93-1.01); 90% CIs were within the predefined range necessary to demonstrate bioequivalence. Hospira filgrastim was well tolerated with no additional safety concerns over Amgen filgrastim. Hospira filgrastim is bioequivalent with Amgen filgrastim with regard to its pharmacodynamic characteristics.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacokinetics , Granulocyte Colony-Stimulating Factor/therapeutic use , Adolescent , Adult , Area Under Curve , Cross-Over Studies , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/adverse effects , Hematologic Diseases/drug therapy , Humans , Male , Middle Aged , Neoplasms/drug therapy , Recombinant Proteins , Therapeutic Equivalency , Young Adult
16.
Ann Hematol ; 89(9): 927-33, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20428872

ABSTRACT

Recombinant human granulocyte colony-stimulating factor (filgrastim) has multiple hematologic and oncologic indications as Neupogen (Amgen filgrastim). Hospira has developed a biosimilar filgrastim (Nivestim). Here, results are reported from a phase I trial, primarily designed to compare the pharmacokinetic profiles of Hospira filgrastim and Amgen filgrastim. A phase I, single-center, open-label, randomized trial was undertaken to demonstrate equivalence of the pharmacokinetic characteristics of Hospira filgrastim and Amgen filgrastim. Forty-eight healthy volunteers were randomized to receive intravenous (i.v.) or subcutaneous (s.c.) dosing and then further randomized to order of treatment. Volunteers in each of the two dosing groups received a single 10microg/kg dose of Hospira filgrastim or Amgen filgrastim, with subsequent crossover. Bioequivalence was evaluated by analysis of variance; if the estimated 90% confidence intervals (CIs) for the ratio of 'test' to 'reference' treatment means were within the conventional equivalence limits of 0.80-1.25, then bioequivalence was concluded. Forty-six volunteers completed the study. Geometric mean area under the curve from time 0 to the last time point (primary endpoint) was similar in volunteers given Hospira filgrastim or Amgen filgrastim following i.v. (ratio of means: 0.96; 90% CI: 0.90-1.02) or s.c. (ratio of means: 1.02; 90% CI: 0.95-1.09) dosing; 90% CIs were within the predefined range necessary to demonstrate bioequivalence. Hospira filgrastim was well tolerated with no additional safety concerns over Amgen filgrastim. Hospira filgrastim is bioequivalent with Amgen filgrastim in terms of its pharmacokinetic properties and may provide a clinically effective alternative.


Subject(s)
Granulocyte Colony-Stimulating Factor/chemistry , Granulocyte Colony-Stimulating Factor/pharmacokinetics , Adolescent , Adult , Chemistry, Pharmaceutical , Cross-Over Studies , Female , Filgrastim , Humans , Male , Middle Aged , Recombinant Proteins , Therapeutic Equivalency , Young Adult
17.
Hum Vaccin ; 5(9): 608-13, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19617717

ABSTRACT

In response to increased demand for the 23-valent pneumococcal vaccine PNEUMOVAX II, a new manufacturing process has been implemented that improves the consistency and increases the scale of production. This double-blind, randomized, clinical study compared the immunogenicity and safety profiles of the new-process PNEUMOVAX II (n = 111) formulated with all 23 new process polysaccharides to the former-process PNEUMOVAX II (n = 109) formulation in adults aged > or =50 years. The primary aim of the study was to compare the post-vaccination geometric mean of antibody titres (GMT) to pneumococcal serotypes 3 and 8 in recipients of new- and former-process PNEUMOVAX II. The post-vaccination GMTs for serotypes 3 and 8 elicited by the new-process PNEUMOVAX II [1.40 (95% confidence interval, 1.21-1.63) and 10.78 (95% CI, 9.10-12.77), respectively] were non-inferior to those elicited by the former-process PNEUMOVAX II [1.24 (95% CI, 1.07-1.43) and 9.72 (95% CI, 8.22-11.50), respectively]. Both PNEUMOVAX II formulations were well tolerated; there were no vaccine-related serious adverse events. A total of 74 (66.7%) subjects in the new-process group and 66 (60.6%) in the former-process group had at least one injection-site reaction or vaccine-related systemic event within 14 days following vaccination. There was a trend for higher incidence of two solicited injection-site reactions (erythmea and induration) and three solicited systemic events (asthenia, chills and body aches) with the new- versus former-process PNEUMOVAX II, but these were mainly of mild intensity and short duration. The new-process PNEUMOVAX(R II thus showed similar immunogenicity to the former-process vaccine for pneumococcal serotypes 3 and 8 and both vaccines were well tolerated in older subjects.


Subject(s)
Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/adverse effects , Pneumococcal Vaccines/immunology , Aged , Aged, 80 and over , Antibodies, Bacterial/blood , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions , Female , Fever/chemically induced , Humans , Male , Middle Aged , Pain/chemically induced , Skin Diseases/chemically induced , Skin Diseases/pathology , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/standards
SELECTION OF CITATIONS
SEARCH DETAIL
...