Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Bioanalysis ; 11(24): 2283-2296, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31845602

ABSTRACT

Therapeutic proteins have the potential to induce unwanted immune responses. The potential impact of immunogenicity on pharmacokinetics, pharmacodynamics, safety and efficacy are well established. Here, we analyze key aspects of current US FDA and EMA guidelines on the development and validation of antidrug antibody assays. Although FDA and EMA guidance documents are in harmony on most points, EMA allows greater leeway for scientific judgement, while FDA recommends specific approaches that may not be appropriate in some situations. Many white papers suggest approaches different from the guidance documents, however, these can conflict with each other and are themselves only scientifically valid in certain situations. Here, we indicate when alternatives to guidance may be needed and what those approaches might be.


Subject(s)
Antibodies/therapeutic use , Proteins/therapeutic use , United States Food and Drug Administration/standards , Antibodies/pharmacology , Humans , Proteins/pharmacology , United States
2.
J Immunol Methods ; 474: 112669, 2019 11.
Article in English | MEDLINE | ID: mdl-31614128

ABSTRACT

Detection of anti-drug antibodies is a critical step in the development of large molecule biopharmaceuticals. In the case of multicomponent/multifunctional molecules, such as fusion proteins and protein conjugates such as covalent polyethylene glycol (PEG)~protein conjugates, it is useful to further characterize anti-drug antibody (ADA) binding to key domains of the drug. The detection of anti-PEG antibodies poses special challenges that if overlooked can result in underreporting antibody responses. Here we describe the development and characterization of a novel ELISA to detect anti-PEG antibodies that provides a more complete interpretation of anti-PEG than other published methods. Being specific to the PEG moiety alone, this method is intended to detect anti-PEG antibodies independent of the protein to which PEG is conjugated. Based upon early indications that our assay could detect anti-PEG antibodies at a surprisingly high frequency in the general population, our emphasis throughout method development and validation was to ensure that non-specific signals and unintended interactions were not falsely contributing to detection of anti-PEG antibodies. Techniques, including orthogonal methods used to ensure that this ELISA detected antibodies specific to PEG included competition, immunodepletion, immunoprecipitation/western blot and an Octet kinetic binding analysis. The validated ELISA can detect 100 ng/mL of an anti-PEG IgG positive control and 800 ng/mL of an anti-PEG IgM positive control in the presence of 7.5 µg/mL of the PEGylated therapeutic (MW 64 kDa). The intra-assay percent co-efficient of variation (CV) and inter-assay CV of the low positive control samples in the screening method were 4.1 to 7.2% and 16.7 to 17.7%, respectively. Additional assay performance parameters that were validated are also described. When the validated assay was applied to a population of 200 healthy blood donors with no known exposure to biopharmaceutical PEG conjugates it indicated a pre-existing anti-PEG antibody prevalence of 97.5%. We suggest this surprising result is a consequence of exposure to PEG additives in everyday products, such as cosmetics, processed foods and over-the-counter (OTC) pharmaceuticals.


Subject(s)
Antigens/immunology , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G/blood , Immunoglobulin M/blood , Polyethylene Glycols , Antigens/metabolism , Binding Sites, Antibody , Humans , Kinetics , Polyethylene Glycols/metabolism , Protein Binding , Reproducibility of Results , Surface-Active Agents/chemistry
3.
Biochem Biophys Res Commun ; 497(2): 467-472, 2018 03 04.
Article in English | MEDLINE | ID: mdl-29470981

ABSTRACT

GhoT is a bacterial toxin of the type V toxin/antitoxin system that allows Escherichia coli to reduce its metabolism in response to oxidative and bile stress. GhoT functions by increasing membrane permeability and reducing both ATP levels and the proton motive force. However, how GhoT damages the inner membrane has not been elucidated. Here we investigated how GhoT damages membranes by studying its interaction with lipid bilayers and determined that GhoT does not cause macroscopic disruption of the lipid bilayer to increase membrane permeability to the dye carboxyfluorescein. Using circular dichroism, we found that GhoT forms an alpha helical structure in lipid bilayers that agrees with the structure predicted by the I-TASSER protein structure prediction program. The structure generated using I-TASSER was used to conduct coarse-grained molecular dynamics simulations, which indicate that GhoT damages the cell membrane, as a multimer, by forming transient transmembrane pores.


Subject(s)
Cell Membrane/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Lipid Bilayers/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Escherichia coli/chemistry , Escherichia coli Infections/pathology , Escherichia coli Proteins/chemistry , Host-Pathogen Interactions , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Multimerization
4.
Bioanalysis ; 9(18): 1407-1422, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28920457

ABSTRACT

Bioanalytical methods must enable the delivery of data that meet sound, scientifically justified, fit-for-purpose criteria. At early phases of biotherapeutic drug development, suitable criteria of a ligand-binding assay could be met for pharmacokinetic (PK) in-study sample testing without a full validation defined by regulatory guidelines. To ensure fit-for-purpose methods support PK testing through all phases of biotherapeutic development, three tiers of method validation - regulatory, scientific and research validations - are proposed. The three-tiered framework for method validation outlines the differences in the parameters that should be assessed, the acceptance criteria that may be applied, and the documentation necessary at each level. The criteria for selecting the appropriate application of each of these PK method validation workflows are discussed.


Subject(s)
Chemistry Techniques, Analytical/methods , Humans , Ligands , Linear Models , Reproducibility of Results , Social Control, Formal , Tissue Distribution
5.
Eur J Clin Pharmacol ; 72(11): 1303-1310, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27515978

ABSTRACT

PURPOSE: The purpose of this study is to evaluate the pharmacokinetics, immunogenicity, safety, and tolerability of guselkumab, a human monoclonal antibody with high affinity and specificity for binding to interleukin-23. METHODS: In this first-in-human, phase 1, randomized study, a single intravenous (IV; 0.03-10 mg/kg) or subcutaneous (SC; 10-300 mg) dose of guselkumab was administered to 47 healthy subjects, and a single SC dose (placebo, 10, 30, 100, 300 mg) was administered to 24 patients with moderate-to-severe psoriasis. RESULTS: Mean maximum observed serum concentration and area under the zero-to-infinity serum concentration-time curve of guselkumab increased in an approximately dose-proportional manner over the dose range of 0.03-10 mg/kg following a single IV administration or 10-300 mg following a single SC administration. Mean clearance and volume of distribution ranged from 3.62-6.03 mL/day/kg and 99.38-123.22 mL/kg, respectively. Mean half-life ranged from 12 to 19 days in healthy subjects and patients with psoriasis. Among guselkumab-treated subjects/patients, 1/30 (3.3 %) healthy subjects in the IV group, 0/6 healthy subjects in the SC group, and 1/20 (5.0 %) patients with psoriasis tested positive for antibodies to guselkumab. No clinically significant adverse events were identified in this study. CONCLUSION: Guselkumab pharmacokinetic profiles were generally comparable between healthy subjects and patients with psoriasis. Guselkumab, administered as an IV infusion or SC injection, was well tolerated in healthy subjects and patients with psoriasis.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Dermatologic Agents/pharmacokinetics , Adolescent , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/blood , Antibodies, Monoclonal, Humanized , Area Under Curve , Dermatologic Agents/administration & dosage , Dermatologic Agents/adverse effects , Dermatologic Agents/blood , Double-Blind Method , Female , Half-Life , Healthy Volunteers , Humans , Infusions, Intravenous , Injections, Subcutaneous , Interleukin-23/antagonists & inhibitors , Interleukin-23/immunology , Male , Middle Aged , Psoriasis/blood , Psoriasis/drug therapy , Psoriasis/metabolism , Young Adult
6.
Langmuir ; 31(15): 4496-502, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25845029

ABSTRACT

A cationic protein isolated from the seeds of the Moringa oleifera tree has been extensively studied for use in water treatment in developing countries and has been proposed for use in antimicrobial and therapeutic applications. However, the molecular basis for the antimicrobial action of this peptide, Moringa oleifera cationic protein (MOCP), has not been previously elucidated. We demonstrate here that a dominant mechanism of MOCP antimicrobial activity is membrane fusion. We used a combination of cryogenic electron microscopy (cryo-EM) and fluorescence assays to observe and study the kinetics of fusion of membranes in liposomes representing model microbial cells. We also conducted cryo-EM experiments on E. coli cells where MOCP was seen to fuse the inner and outer membranes. Coarse-grained molecular dynamics simulations of membrane vesicles with MOCP molecules were used to elucidate steps in peptide adsorption, stalk formation, and fusion between membranes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Escherichia coli/drug effects , Moringa oleifera/chemistry , Plant Proteins/pharmacology , Seeds/chemistry , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Cations , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cryoelectron Microscopy , Escherichia coli/chemistry , Escherichia coli/ultrastructure , Flocculation , Fresh Water/microbiology , Kinetics , Liposomes/chemistry , Liposomes/ultrastructure , Membrane Fusion/drug effects , Molecular Dynamics Simulation , Molecular Sequence Data , Plant Extracts/chemistry , Plant Proteins/chemistry , Plant Proteins/isolation & purification , Protein Structure, Secondary , Spectrometry, Fluorescence , Water Purification/methods
7.
AAPS J ; 16(3): 464-77, 2014 May.
Article in English | MEDLINE | ID: mdl-24590506

ABSTRACT

Fulranumab, a human IgG2 monoclonal antibody that neutralizes nerve growth factor (NGF), is currently in development for the treatment of pain. Our initial immunogenicity test method was found to be prone to NGF interference, leading to a high apparent incidence of anti-drug antibody (ADA) in phase 1 studies. The ADA immunoassay comprised a homogeneous bridging electrochemiluminescence (ECL) format with biotin and ruthenium-labeled fulranumab bound together ("bridged") by ADA in test samples for detection. In this assay, NGF produced a false-positive signal due to its ability to bridge fulranumab molecules. Thus, we developed a specificity assay to eliminate the NGF false-positive results. We encountered the challenge of eliminating drug interference as well as drug target interference, and discovered that the acid-dissociation-based pretreatment of samples used for mitigating drug interference dramatically increased drug target interference. Several strategies were investigated to eliminate the NGF interference; yet only one strategy specifically removed NGF and produced true fulranumab-specific ADA results by using competitive inhibition with fulranumab and utilizing an alternative NGF binding antibody to eliminate NGF interference. Using this new method, we confirmed that the high apparent anti-fulranumab antibody incidence (>60%) in clinical study samples was in fact due to fulranumab-bound NGF released during the acid-dissociation step of the ADA testing method. We conclude that our revised method accurately identifies anti-fulranumab antibodies by incorporating steps to eliminate fulranumab and NGF interference. We advise that acid-dissociation pretreatment must not be universally applied to improve ADA assays without investigating its bioanalytical risks versus benefits.


Subject(s)
Antibodies, Monoclonal/immunology , Nerve Growth Factor/chemistry , Antibodies, Blocking/chemistry , Antibodies, Immobilized/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , False Positive Reactions , Humans , Immunoglobulin G/analysis , Immunoglobulin G/immunology , Nerve Growth Factor/isolation & purification
8.
J Pharm Biomed Anal ; 54(2): 286-94, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-20869832

ABSTRACT

We assessed the utility of the FortéBio Octet(®) system for detection of anti-drug antibodies (ADAs) against an investigational therapeutic human IgG1 monoclonal antibody (mAb), CNTO X. To understand the relative merits of this technology, key performance requirements were compared with two popularly accepted ADA detection methods, a step-wise bridging ELISA and a Meso Scale Discovery (MSD) homogeneous (single step binding) bridging ECLIA. When used to detect 13 monoclonal ADAs of varying affinities and one polyclonal ADA, all three methods demonstrated their greatest apparent sensitivity to the polyclonal sample (1, 6, and 130 ng/mL, respectively for ECLIA, ELISA, and Octet). Sensitivity to monoclonal ADAs tended to vary in accordance with their affinities, however, the sensitivity of the Octet method varied much less between ADAs. As a result, the above ranking became reversed such that Octet was the most and ELISA least sensitive for detection of low-affinity ADAs. With regard to drug tolerance, the presence of CNTO X could lead to false-negative assay results, although each method was affected to a different degree, with the Octet method tolerating up to 10 times more drug than the ECLIA method, which in turn tolerated up to 10 times more than the ELISA. Finally, the ECLIA and Octet methods were applied to the bioanalysis of cynomolgus monkey sera from a pre-clinical multiple dose study of CNTO X. Octet indicated 3 positive animals developed ADA as early as day 15 of the dosing phase while drug was present at nearly 1mg/mL. ECLIA detected only one of these, and only in a day 57 recovery sample after drug had cleared from circulation. We conclude that the Octet is a promising platform for detection of lower affinity ADAs and is particularly suitable for ADA detection when drug persists at levels that negatively impact bridging immunoassays.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Drug Tolerance/immunology , Interferometry/methods , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antibody Affinity , Antibody Formation , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay/methods , False Negative Reactions , Female , Humans , Immunoassay/methods , Immunoglobulin G , Limit of Detection , Luminescent Measurements/methods , Macaca fascicularis/immunology , Macaca fascicularis/metabolism , Sensitivity and Specificity , Serum/chemistry , Serum/metabolism
9.
Birth Defects Res B Dev Reprod Toxicol ; 89(2): 116-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20151458

ABSTRACT

BACKGROUND: Intetumumab is a human IgG1 anti-alphav-integrin monoclonal antibody that inhibits angiogenesis. Integrin binding and angiogenesis are important in reproduction including fertilization, implantation, and embryofetal development. These studies were designed to determine the pharmacological relevance of the rabbit for the evaluation of potential effects on embryofetal development and to evaluate the placental transfer of intetumumab in rabbits. METHODS: In vitro pharmacology studies evaluated the binding of intetumumab to rabbit cells and the inhibition of vessel sprouting from rabbit aorta. For the evaluation of placental transfer, pregnant rabbits (8/group) were injected intravenously with intetumumab 50 or 100 mg/kg every 2 days from Gestation Day (GD)7 to GD19. Maternal sera, fetal homogenates/sera, and amniotic fluid were collected at necropsy on GD19 or GD28 for evaluation of intetumumab concentrations. Clinical condition of the dams was monitored and fetuses were screened for abnormalities. RESULTS: Intetumumab (5-40 microg/mL) inhibited aortic cell adhesion to vitronectin and vessel sprouting from rabbit aortic rings. Immunohistochemical staining of rabbit tissues demonstrated binding of intetumumab to placenta. Administration of intetumumab to pregnant rabbits was well tolerated by the dams and the fetuses did not show major abnormalities. Fetal exposure to intetumumab relative to maternal exposure was <0.1% on GD19 and 100-130% on GD29. CONCLUSIONS: The rabbit is a pharmacologically relevant species for evaluation of potential developmental effects of intetumumab. Intetumumab crosses the rabbit placenta during the fetal period (GD 19-28).


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Integrin alpha5/immunology , Maternal-Fetal Exchange/drug effects , Placenta/drug effects , Amniotic Fluid/drug effects , Amniotic Fluid/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Blood Vessels/drug effects , Blood Vessels/growth & development , Cell Adhesion/drug effects , Cells, Cultured , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Embryonic Development/physiology , Endothelial Cells/drug effects , Female , Fetal Development/drug effects , Fetal Development/physiology , Fetus/drug effects , Fetus/metabolism , Injections, Intravenous , Maternal Exposure , Maternal-Fetal Exchange/physiology , Neovascularization, Physiologic/drug effects , Placenta/metabolism , Pregnancy , Rabbits
10.
Cell Immunol ; 253(1-2): 31-7, 2008.
Article in English | MEDLINE | ID: mdl-18501882

ABSTRACT

The T-cell cytokine IL-17 is implicated in multiple inflammatory diseases through its induction of several pro-inflammatory cytokines and chemokines in a broad range of cell targets. Production of IL-17 defines the Th17 subset of helper T-cells associated with protection against microorganisms, a profile best characterized in the murine system. Multiple regulators of Th17 cell differentiation and IL-17 production are reported, but the impact of OX40L is not described. OX40 ligand (OX40L) is an early-stage activator of T-cells through its interaction with CD134 (OX40) that is up-regulated on antigen challenged T-cells. Here, we show that OX40L suppresses IL-17 production by PHA-stimulated human PBMC and purified CD4 and CD8 cells. In agreement with prior reports, OX40L signaling through CD134 increased IFNgamma and IL-4, both of which are reported to inhibit the production of IL-17. OX40L suppression of IL-17 was completely reversed by a neutralizing IFNgamma antibody while there was no effect with a neutralizing IL-4 antibody. Moreover, OX40L also suppressed IL-17 in the presence of IL-23, an established inducer of IL-17 and differentiation factor for Th17 cells. Presuming mediation by IFNgamma, we evaluated expression of this cytokine in the presence of OX40L and IL-23. Surprisingly, IL-23 also induced IFNgamma by PHA-stimulated T-cells and this effect was enhanced in the presence of OX40L. Addition of the IFNgamma antibody not only reversed the OX40L suppression of IL-17 in the presence of IL-23, it markedly enhanced the level of IL-17. These results further establish IFNgamma as a primary modulator of IL-17 production in the human cells, much as in the murine system.


Subject(s)
Interleukin-17/immunology , OX40 Ligand/immunology , Receptors, OX40/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Humans , Interferon-gamma/immunology , Interleukin-23/immunology , Interleukin-4/immunology , Interleukin-6/immunology , Mice , Phytohemagglutinins/immunology
11.
Clin Cancer Res ; 11(19 Pt 1): 6959-65, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16203788

ABSTRACT

PURPOSE: CNTO 95 is a fully human anti-alphav integrin monoclonal antibody that inhibits macaque and rodent angiogenesis and inhibits human tumor growth in rodents. The purpose of these studies was to evaluate the preclinical safety of long-term administration of CNTO 95 in cynomolgus macaques. EXPERIMENTAL DESIGN: The in vitro binding profiles of CNTO 95 to human and macaque tissues and the in vivo binding to macaque tissues was evaluated by immunohistochemistry. The preclinical safety of CNTO 95 (10 and 50 mg/kg, i.v.) was evaluated in macaques treated once per week for up to 6 months. Safety was evaluated by clinical observations, ophthalmic and physical examinations (including heart rate, blood pressure, and electrocardiogram), clinical pathology (including coagulation parameters), and comprehensive anatomic pathology. The effect of CNTO 95 (50 mg/kg, i.v.) on incisional wound healing was evaluated in macaques. RESULTS: The tissue binding studies showed that CNTO 95 bound with mild to moderate intensity to macaque and human endothelial cells, epithelial cells, and vascular smooth muscle cells in most normal tissues examined. CNTO 95 showed strong to intense staining to the positive control tissue, human placenta. Despite the widespread binding to normal tissues, treatment of cynomolgus macaques with CNTO 95 produced no signs of toxicity and no histopathologic changes in any of the tissues examined (including ovaries and bone growth plates). CNTO 95 did not impair wound healing. CONCLUSION: These studies show that CNTO 95 is safe and, unlike some other angiogenesis inhibitors, does not seem to inhibit normal physiologic angiogenesis.


Subject(s)
Antibodies, Monoclonal/chemistry , Integrin alphaV/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Aorta/metabolism , Area Under Curve , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Female , Humans , Immune System , Immunohistochemistry , Macaca fascicularis , Male , Myocytes, Smooth Muscle/cytology , Neovascularization, Pathologic , Placenta/metabolism , Protein Binding , Time Factors , Wound Healing
12.
Cancer Immunol Immunother ; 54(11): 1082-94, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16047142

ABSTRACT

Prostate specific antigen (PSA) is a serum marker that is widely used in the detection and monitoring of prostate cancer. Though PSA is a self-antigen, T cell responses to PSA epitopes have been detected in healthy men and prostate cancer patients, suggesting it may be used as a target for active immunotherapy of prostate cancer. A PSA DNA vaccine (pPSA) was evaluated in mice and monkeys for its ability to induce antigen-specific immune responses. Mice immunized intradermally with pPSA demonstrated strong PSA-specific humoral and cellular immunity. The anti-PSA immune responses were skewed toward Th1, as shown by high IFNgamma and IL-2 production. The immune response was sufficient to protect mice from challenge with PSA-expressing tumor cells. Tumor protection was durable in the absence of additional vaccination, as demonstrated by protection of vaccinated mice from tumor rechallenge. Furthermore, pPSA vaccination induced PSA-specific antibody titers in male cynomolgus monkeys, which express a closely related PSA gene. These results demonstrate that vaccination with pPSA may be able to break tolerance and can induce an immune response that mediates tumor protection.


Subject(s)
Prostate-Specific Antigen/immunology , Prostatic Neoplasms/prevention & control , Th1 Cells/immunology , Vaccines, DNA/immunology , Animals , Antibodies/blood , Female , Humans , Immunization , Interferon-gamma/biosynthesis , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/immunology
13.
J Pharm Biomed Anal ; 39(3-4): 364-75, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15963677

ABSTRACT

Immunogenicity has always been an important consideration in the evaluation of pharmaceutical protein biologics. In this article, method validation parameters relevant to enzyme immunoassays are described for assays applied to the analysis of anti-drug antibodies, with special considerations for immunogenicity to therapeutic monoclonal antibodies. Common strategies for experimental investigation of various validation parameters are proposed. In addition, a novel, yet simple, approach is proposed to categorize the validation effort into two mutually interdependent phases, based on the characterization of validation parameters as "system descriptive" or "system controlled". System descriptive parameters are those that must be characterized but need not have pre-specified acceptance criteria for assay validation. In contrast, system-controlled parameters should be understood early in assay development, and optimized and confirmed using a priori acceptance criteria in validation to assure sufficient control over them during routine bioanalysis. This approach not only streamlines the validation process but also eliminates unnecessary redundancies. This validation method can be achieved with proper scientific rigor and remain within the realm of GLP compliance. The authors hope that other research groups would engage in discussions on validation of anti-drug antibody assays in order to establish a consistent approach across the industry and academia.


Subject(s)
Antibodies, Monoclonal/chemistry , Drug Industry/methods , Immunoassay/methods , Animals , Chemistry, Pharmaceutical/methods , Enzyme-Linked Immunosorbent Assay , Humans , Immunoenzyme Techniques/methods , Pharmaceutical Preparations/chemistry , Reproducibility of Results , Sensitivity and Specificity
14.
Cell Immunol ; 227(2): 103-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15135292

ABSTRACT

IL-18 is a pluripotent proinflammatory cytokine produced primarily by antigen presenting cells involved in numerous aspects of immune regulation most notably on lymphoid cells. The effect of IL-18 stimulation on cells in the myeloid compartment, however, has been poorly studied. Human monocytes did not respond to IL-18. However, the human myelomonocytic cell line KG-1 and monocyte-derived dendritic cells (generated by GM-CSF+IL-4) showed a marked increase in CD83, HLA-DR, and several costimulatory molecules upon stimulation with IL-18. Furthermore, IL-18 decreased pinocytosis of these cells and increased their ability to stimulate alloreactive T cell proliferation, all characteristics of mature dendritic cells. These results suggest that IL-18 is involved in the maturation of myeloid DCs, but not differentiation of monocytes into DCs. The finding that IL-18 is involved in the maturation of dendritic cells is both novel and unexpected and indicates another important role for IL-18 as a key regulator of immune responses.


Subject(s)
Dendritic Cells/drug effects , Interleukin-18/pharmacology , Cell Line , Dendritic Cells/physiology , Endocytosis/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-18 Receptor alpha Subunit , Lymphocyte Culture Test, Mixed , Receptors, Interleukin/genetics , Receptors, Interleukin-18 , Up-Regulation
15.
Curr Opin Mol Ther ; 5(2): 172-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12772508

ABSTRACT

There are currently 13 monoclonal antibodies or antibody constructs approved for therapeutic use in the US and at least another 400 products are in clinical testing or undergoing regulatory review. Despite widespread use and development of therapeutic monoclonal antibodies, limited information exists regarding the incidence and consequence of immune antibodies generated against many of these products. In this review, analytical methodology and recent data on the immunogenicity of approved therapeutic monoclonal antibodies will be presented. The implications of these findings in relation to potential concerns for repeated or chronic treatment with these powerful and specific therapeutic agents will be discussed.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Immunity , Animals , Antibodies, Monoclonal/pharmacokinetics , Crohn Disease/drug therapy , Crohn Disease/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunity/drug effects , Infliximab , Radioimmunoassay , Surface Plasmon Resonance
16.
J Immunol ; 169(11): 6554-63, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12444167

ABSTRACT

The experimental autoimmune encephalomyelitis (EAE) model in the common marmoset approximates recognized features of the human disease multiple sclerosis (MS) with regard to its clinical presentation as well as neuropathological and radiological aspects of the lesions in brain and spinal cord. IL-12 is a proinflammatory cytokine that is produced by APC and promotes differentiation of Th1 effector cells. IL-12 is produced in the developing lesions of patients with MS as well as in EAE-affected animals. Previously it was shown that interference in IL-12 pathways effectively prevents EAE in rodents. In this study we report that in vivo neutralization of IL-12p40 using a novel Ab has beneficial effects in the myelin-induced EAE model in common marmosets. The Ab was injected i.v. at 7-day intervals starting well after immunization (day 14) and was continued until the end of the study (day 86). Stable levels of the Ab were measured 3 days after each injection throughout the study period. During this period anti-Ab responses could not be detected. We demonstrate that anti-IL-12p40 treatment has a protective effect on the neurological dysfunction as well as on neuropathological changes normally observed in the brain and spinal cord of EAE-affected individuals.


Subject(s)
Antibodies, Monoclonal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interleukin-12/antagonists & inhibitors , Protein Subunits/antagonists & inhibitors , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Monoclonal/administration & dosage , B-Lymphocytes/immunology , Brain/immunology , Brain/pathology , Callithrix , Cross Reactions , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Interleukin-12 Subunit p40 , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Myelin Proteins/immunology , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...