Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
PLoS One ; 16(3): e0247972, 2021.
Article in English | MEDLINE | ID: mdl-33684160

ABSTRACT

INTRODUCTION: Delayed graft function (DGF) following renal transplantation is a manifestation of acute kidney injury (AKI) leading to poor long-term outcome. Current treatments have limited effectiveness in preventing DGF. Interleukin-18 (IL18), a biomarker of AKI, induces interferon-γ expression and immune activation. GSK1070806, an anti-IL18 monoclonal antibody, neutralizes activated (mature) IL18 released from damaged cells following inflammasome activation. This phase IIa, single-arm trial assessed the effect of a single dose of GSK1070806 on DGF occurrence post donation after circulatory death (DCD) kidney transplantation. METHODS: The 3 mg/kg intravenous dose was selected based on prior studies and physiologically based pharmacokinetic (PBPK) modeling, indicating the high likelihood of a rapid and high level of IL18 target engagement when administered prior to kidney allograft reperfusion. Utilization of a Bayesian sequential design with a background standard-of-care DGF rate of 50% based on literature, and confirmed via extensive registry data analyses, enabled a statistical efficacy assessment with a minimal sample size. The primary endpoint was DGF frequency, defined as dialysis requirement ≤7 days post transplantation (except for hyperkalemia). Secondary endpoints included safety, pharmacokinetics and pharmacodynamic biomarkers. RESULTS: GSK1070806 administration was associated with IL18-GSK1070806 complex detection and increased total serum IL18 levels due to IL18 half-life prolongation induced by GSK1070806 binding. Interferon-γ-induced chemokine levels declined or remained unchanged in most patients. Although the study was concluded prior to the Bayesian-defined stopping point, 4/7 enrolled patients (57%) had DGF, exceeding the 50% standard-of-care rate, and an additional two patients, although not reaching the protocol-defined DGF definition, demonstrated poor graft function. Six of seven patients experienced serious adverse events (SAEs), including two treatment-related SAEs. CONCLUSION: Overall, using a Bayesian design and extensive PBPK dose modeling with only a small sample size, it was deemed unlikely that GSK1070806 would be efficacious in preventing DGF in the enrolled DCD transplant population. TRIAL REGISTRATION: NCT02723786.


Subject(s)
Acute Kidney Injury , Antibodies, Monoclonal, Humanized , Delayed Graft Function , Interleukin-18/blood , Kidney Transplantation , Tissue Donors , Acute Kidney Injury/blood , Acute Kidney Injury/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Delayed Graft Function/blood , Delayed Graft Function/drug therapy , Female , Humans , Male , Middle Aged , Pilot Projects
2.
Clin Exp Immunol ; 192(3): 348-365, 2018 06.
Article in English | MEDLINE | ID: mdl-29431871

ABSTRACT

We examined the assay formats used to detect anti-drug antibodies (ADA) in clinical studies of the anti-tumour necrosis factor (TNF) monoclonal antibodies adalimumab and infliximab in chronic inflammatory disease and their potential impact on pharmacokinetic and clinical outcomes. Using findings of a recent systematic literature review of the immunogenicity of 11 biological/biosimilar agents, we conducted an ancillary qualitative review of a subset of randomized controlled trials and observational studies of the monoclonal antibodies against anti-TNF factor adalimumab and infliximab. Among studies of adalimumab and infliximab, the immunoassay method used to detect antibodies was reported in 91 of 111 (82%) and 154 of 206 (75%) adalimumab and infliximab studies, respectively. In most adalimumab and infliximab studies, an enzyme-linked immunosorbent assay or radioimmunoassay was used [85 of 91 (93%) and 134 of 154 (87%), respectively]. ADA incidence varied widely among assays and inflammatory diseases (adalimumab, 0-87%; infliximab, 0-79%). Pharmacokinetic and clinical outcomes were only reported for ADA-positive patients in 38 of 91 (42%) and 61 of 154 (40%) adalimumab and infliximab studies, respectively. Regardless of assay format or biological used, ADA formation was associated with lower serum concentrations, reduced efficacy and elevated rates of infusion-related reactions. Consistent with previous recommendations to improve interpretation of immunogenicity data for biologicals, greater consistency in reporting of assay methods and clinical consequences of ADA formation may prove useful. Additional standardization in immunogenicity testing and reporting, application of modern, robust assays that satisfy current regulatory expectations and implementation of international standards for marketed products may help to improve our understanding of the impact of immunogenicity to biologics.


Subject(s)
Adalimumab/immunology , Antibodies/immunology , Antirheumatic Agents/immunology , Enzyme-Linked Immunosorbent Assay/methods , Infliximab/immunology , Radioimmunoassay/methods , Adalimumab/therapeutic use , Antirheumatic Agents/therapeutic use , Humans , Infliximab/therapeutic use , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
3.
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
4.
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
5.
Cell Growth Differ ; 10(11): 739-48, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10593650

ABSTRACT

By crossing TG.AC v-Ha-ras and K6/ODC transgenic mice, we found previously that an activated ras and follicular ornithine decarboxylase (ODC) overexpression cooperate to generate spontaneous tumors in the skin. Cellular proliferation was dramatically increased in the K6/ODC transgenic skin, as evidenced by elevated proliferating cell nuclear antigen and Ki67 expression compared with nontransgenic littermates. Keratinocytes isolated from transgenic skin also displayed increased clonal growth. Paradoxically, expression of the growth inhibition-associated proteins p53, p21Waf1, p27Klp1, and Bax was increased with ODC overexpression in the skin. ODC overexpression did not affect cyclin D/cyclin-dependent kinase 4 (Cdk4)-dependent phosphorylation of retinoblastoma protein but stimulated cyclin E/Cdk2 and cyclin A/Cdk2-associated kinase activity, with minimal effect on the levels of these proteins. Thus, ODC/polyamine-induced activation of cyclin E/Cdk2 and cyclin A/Cdk2-associated kinase activity may cooperate with the ras induction of cyclin D/Cdk4/6-associated retinoblastoma protein phosphorylation to not only stimulate proliferation but ultimately contribute to tumor development.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Gene Expression Regulation, Enzymologic , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Skin/cytology , Skin/enzymology , Tumor Suppressor Proteins , Animals , Apoptosis/physiology , Cell Division/physiology , Cell Nucleus/chemistry , Cells, Cultured , Cyclin A/physiology , Cyclin E/physiology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/physiology , Cyclins/metabolism , Enzyme Activation/genetics , Genes, ras/physiology , In Situ Nick-End Labeling , Keratinocytes/cytology , Keratinocytes/enzymology , Ki-67 Antigen/analysis , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Proliferating Cell Nuclear Antigen/analysis , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/metabolism
6.
Nat Biotechnol ; 17(10): 984-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10504699

ABSTRACT

Molecules that selectively target and occlude new blood vessels would be useful for diagnosis and treatment of pathologies associated with angiogenesis. We show that a phage-derived human antibody fragment (L19) with high affinity for the ED-B domain of fibronectin, a marker of angiogenesis, selectively localizes to newly formed blood vessels in a rabbit model of ocular angiogenesis. The L19 antibody, chemically coupled to a photosensitizer and irradiated with red light, mediates complete and selective occlusion of ocular neovasculature and promotes apoptosis of the corresponding endothelial cells. These results demonstrate that new ocular blood vessels can be distinguished immunochemically from preexisting ones and suggest that the targeted delivery of photosensitizers may be effective in treating angiogenesis-related pathologies.


Subject(s)
Eye/blood supply , Immunoglobulin Fragments/immunology , Light Coagulation , Neovascularization, Pathologic/radiotherapy , Animals , Bacteriophages/genetics , Eye/metabolism , Humans , Immunoglobulin Fragments/genetics , Immunohistochemistry , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology
7.
J Immunol Methods ; 231(1-2): 239-48, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10648941

ABSTRACT

Angiogenesis, the formation of new blood vessels from pre-existing ones, is a characteristic process which underlies many diseases, including cancer, rheumatoid arthritis and blinding ocular disorders. Antibodies capable of selective targeting and occlusion of neovasculature would open diagnostic and therapeutic opportunities. We have recently demonstrated that phage-derived human antibody fragments with high affinity for the extra-domain B (ED-B) of fibronectin, a marker of angiogenesis, selectively localise in new-forming blood vessels upon intravenous injection. Here, we show that infrared fluorescence methodologies nicely complement radioactive techniques for the study of the antibody-mediated targeting of angiogenesis in a variety of animal models. Methods are presented for the construction and use of infrared fluorescence imagers, as well as for the production and characterisation of recombinant antibodies labeled with infrared fluorophores.


Subject(s)
Antibodies/analysis , Biomarkers, Tumor/immunology , Neovascularization, Pathologic/immunology , Spectrophotometry, Infrared , Animals , Antibodies/immunology , Chick Embryo , Eye/blood supply , Fibronectins/immunology , Humans , Injections, Intravenous , Mice , Peptide Library , Rabbits , Recombinant Proteins/analysis , Recombinant Proteins/immunology , Spectrometry, Fluorescence , Teratocarcinoma/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...