Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Exp Immunol ; 182(2): 139-48, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26178412

ABSTRACT

During clinical trials of a tumour necrosis factor (TNF)-R1 domain antibody (dAb™) antagonist (GSK1995057), infusion reactions consistent with cytokine release were observed in healthy subjects with high levels of a novel, pre-existing human anti-VH (HAVH) autoantibody. In the presence of HAVH autoantibodies, GSK1995057 induced cytokine release in vitro due to binding of HAVH autoantibodies to a framework region of the dAb. The epitope on GSK1995057 was characterized and dAbs with reduced binding to HAVH autoantibodies were generated; pharmacological comparability was determined in human in-vitro systems and in-vivo animal experiments. A Phase I clinical trial was conducted to investigate the safety and tolerability of the modified dAb (GSK2862277). A significant reduction in HAVH binding was achieved by adding a single alanine residue at the C-terminus to create GSK2862277. Screening a pool of healthy donors demonstrated a reduced frequency of pre-existing autoantibodies from 51% to 7%; in all other respects, GSK2862277 and the parent dAb were comparable. In the Phase I trial, GSK2862277 was well tolerated by both the inhaled and intravenous routes. One subject experienced a mild infusion reaction with cytokine release following intravenous dosing. Subsequently, this subject was found to have high levels of a novel pre-existing antibody specific to the extended C-terminus of GSK2862277. Despite the reduced binding of GSK2862277 to pre-existing HAVH autoantibodies, adverse effects associated with the presence of a novel pre-existing antibody response specific to the modified dAb framework were identified and highlight the challenge of developing biological antagonists to this class of receptor.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Autoantibodies/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Administration, Inhalation , Administration, Intravenous , Adult , Aged , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/blood , Antibody Specificity/immunology , Autoantibodies/blood , Cell Line , Cell Line, Tumor , Epitopes/immunology , Female , Humans , Interleukin-8/immunology , Interleukin-8/metabolism , Macaca fascicularis , Male , Middle Aged , Protein Binding/immunology , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Time Factors , Young Adult
2.
Br J Sports Med ; 47 Suppl 1: i59-69, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24282209

ABSTRACT

OBJECTIVES: To examine with a parallel group study design the performance and physiological responses to a 14-day off-season 'live high-train low in the heat' training camp in elite football players. METHODS: Seventeen professional Australian Rules Football players participated in outdoor football-specific skills (32 ± 1°C, 11.5 h) and indoor strength (23 ± 1°C, 9.3 h) sessions and slept (12 nights) and cycled indoors (4.3 h) in either normal air (NORM, n=8) or normobaric hypoxia (14 ± 1 h/day, FiO2 15.2-14.3%, corresponding to a simulated altitude of 2500-3000 m, hypoxic (HYP), n=9). They completed the Yo-Yo Intermittent Recovery level 2 (Yo-YoIR2) in temperate conditions (23 ± 1°C, normal air) precamp (Pre) and postcamp (Post). Plasma volume (PV) and haemoglobin mass (Hb(mass)) were measured at similar times and 4 weeks postcamp (4WPost). Sweat sodium concentration ((Na(+))(sweat)) was measured Pre and Post during a heat-response test (44°C). RESULTS: Both groups showed very large improvements in Yo-YoIR2 at Post (+44%; 90% CL 38, 50), with no between-group differences in the changes (-1%; -9, 9). Postcamp, large changes in PV (+5.6%; -1.8, 5.6) and (Na(+))sweat (-29%; -37, -19) were observed in both groups, while Hb(mass) only moderately increased in HYP (+2.6%; 0.5, 4.5). At 4WPost, there was a likely slightly greater increase in Hb(mass) (+4.6%; 0.0, 9.3) and PV (+6%; -5, 18, unclear) in HYP than in NORM. CONCLUSIONS: The combination of heat and hypoxic exposure during sleep/training might offer a promising 'conditioning cocktail' in team sports.


Subject(s)
Altitude , Football/physiology , Acclimatization/physiology , Athletic Performance/physiology , Atmospheric Pressure , Australia , Blood Volume/physiology , Exercise/physiology , Hemoglobins/metabolism , Hot Temperature , Humans , Hypoxia/physiopathology , Running/physiology , Sleep/physiology
3.
J Clin Immunol ; 33(7): 1192-203, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23832582

ABSTRACT

PURPOSE: To investigate the impact of a new class of anti-Ig autoantibodies reactive with variable heavy (VH) chain framework sequences (human anti-VH autoantibodies) on the pharmacology and safety of an anti-TNFR1 VH domain antibody (GSK1995057) in healthy human subjects. METHODS: Single-blind, randomised, placebo-controlled dose escalation study in which healthy males (n = 28) received a single GSK1995057 intravenous infusion of 0.0004, 0.002 and 0.01 mg/kg. All enrolled subjects were pre-screened for human anti-VH (HAVH) autoantibody status and prospectively stratified accordingly. Serum samples from drug-naïve, HAVH-positive volunteers were used to investigate the effect of HAVH/GSK1995057 complexes on the activation of TNFR1 and cytokine release in vitro. RESULTS: Human anti-VH autoantibodies were detected in approximately 50 % of drug-naïve healthy human subjects and clinical and in vitro studies were performed to evaluate their impact on the pharmacology and safety of GSK1995057. We demonstrated that formation of HAVH autoantibody/GSK1995057 complexes activated TNFR1 and caused cytokine release in vitro in some, but not all, of the human cell types tested. When GSK1995057 was administered to healthy subjects, clinical and physiological signs of cytokine release were observed in two HAVH autoantibody-positive subjects following GSK1995057 infusion. In vitro, HAVH autoantibody levels correlated with TNFR1-dependent cytokine release and propensity for cytokine release in humans following GSK1995057 dosing. CONCLUSIONS: Our data support a greater focus on the impact of pre-existing, drug-reactive autoantibodies on the development of antibody fragments and biotherapeutics targeting cell surface receptors.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Autoantibodies/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Adolescent , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Biomarkers, Pharmacological/metabolism , Female , Humans , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Male , Middle Aged , Molecular Targeted Therapy , Pharmacology, Clinical , Prospective Studies , Receptors, Tumor Necrosis Factor, Type I/immunology , Signal Transduction , Young Adult
4.
J Sci Med Sport ; 16(6): 550-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23332540

ABSTRACT

OBJECTIVES: To examine the usefulness of selected physiological and perceptual measures to monitor fitness, fatigue and running performance during a pre-season, 2-week training camp in eighteen professional Australian Rules Football players (21.9±2.0 years). DESIGN: Observational. METHODS: Training load, perceived ratings of wellness (e.g., fatigue, sleep quality) and salivary cortisol were collected daily. Submaximal exercise heart rate (HRex) and a vagal-related heart rate variability index (LnSD1) were also collected at the start of each training session. Yo-Yo Intermittent Recovery level 2 test (Yo-YoIR2, assessed pre-, mid- and post-camp, temperate conditions) and high-speed running distance during standardized drills (HSR, >14.4 km h(-1), 4 times throughout, outdoor) were used as performance measures. RESULTS: There were significant (P<0.001 for all) day-to-day variations in training load (coefficient of variation, CV: 66%), wellness measures (6-18%), HRex (3.3%), LnSD1 (19.0%), but not cortisol (20.0%, P=0.60). While the overall wellness (+0.06, 90% CL (-0.14; 0.02) AU day(-1)) did not change substantially throughout the camp, HRex decreased (-0.51 (-0.58; -0.45)% day(-1)), and cortisol (+0.31 (0.06; 0.57) nmol L(-1)day(-1)), LnSD1 (+0.1 (0.04; 0.06) ms day(-1)), Yo-YoIR2 performance (+23.7 (20.8; 26.6) m day(-1), P<0.001), and HSR (+4.1 (1.5; 6.6) m day(-1), P<0.001) increased. Day-to-day ΔHRex (r=0.80, 90% CL (0.75; 0.85)), ΔLnSD1 (0.51 (r=0.40; 0.62)) and all wellness measures (0.28 (-0.39; -0.17)

Subject(s)
Football/physiology , Physical Conditioning, Human , Fatigue/psychology , Football/psychology , Humans , Male , Psychometrics , Running/physiology , Young Adult
5.
Acta Neurol Scand Suppl ; 185: 63-70, 2006.
Article in English | MEDLINE | ID: mdl-16866913

ABSTRACT

This communication integrates the purported role of cholesterol and statins in Alzheimer's disease (AD) with recent data. Meta-analysis of association studies relevant to AD indicates that apolipoprotein (apo)E4 is the only cholesterol-related polymorphism that shows clear association with AD. This suggests that the effect of apoE4 on the pathophysiology of AD occurs via a mechanism that is not directly related to cholesterol, such as fibrillization of Abeta. Despite the lack of genetic association, cholesterol and statins clearly modulate amyloid precursor protein (APP) processing in cell culture and animal models. Statins appear to act by a pleiotropic mechanism, involving both cholesterol (via lipid rafts) and isoprenylation. The pleiotropic mechanism of statin action clarifies conflicting data from clinical studies, where statins exert an action on Abeta and AD that might be dose dependent because of actions on both cholesterol and isoprenylation. Reduced isoprenylation can also inhibit inflammation. Our own studies of brains from Alzheimer subjects +/- statins indicate that statins inhibit inflammation in humans but might not reduce cerebral Abeta load. These results suggest that the primary action of statins in humans with AD might be to reduce inflammation rather than decrease Abeta load.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/prevention & control , Cholesterol/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Amyloid beta-Protein Precursor/metabolism , Apolipoprotein E4 , Apolipoproteins E/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...