Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
AAPS J ; 19(2): 377-385, 2017 03.
Article in English | MEDLINE | ID: mdl-28083796

ABSTRACT

All biotherapeutics have the potential to induce an immune response. This immunological response is complex and, in addition to antibody formation, involves T cell activation and innate immune responses that could contribute to adverse effects. Integrated immunogenicity data analysis is crucial to understanding the possible clinical consequences of anti-drug antibody (ADA) responses. Because patient- and product-related factors can influence the immunogenicity of a therapeutic protein, a risk-based approach is recommended and followed by most drug developers to provide insight over the potential harm of unwanted ADA responses. This paper examines mitigation strategies currently implemented and novel under investigation approaches used by drug developers. The review describes immunomodulatory regimens used in the clinic to mitigate deleterious ADA responses to replacement therapies for deficiency syndromes, such as hemophilia A and B, and high risk classical infantile Pompe patients (e.g., cyclophosphamide, methotrexate, rituximab); novel in silico and in vitro prediction tools used to select candidates based on their immunogenicity potential (e.g., anti-CD52 antibody primary sequence and IFN beta-1a formulation); in vitro generation of tolerogenic antigen-presenting cells (APCs) to reduce ADA responses to factor VIII and IX in murine models of hemophilia; and selection of novel delivery systems to reduce in vivo ADA responses to highly immunogenic biotherapeutics (e.g., asparaginase). We conclude that mitigation strategies should be considered early in development for biotherapeutics based on our knowledge of existing clinical data for biotherapeutics and the immune response involved in the generation of these ADAs.


Subject(s)
Drug Design , Immunologic Factors/administration & dosage , Proteins/administration & dosage , Animals , Antibodies/immunology , Antibody Formation/immunology , Antigen-Presenting Cells/immunology , Disease Models, Animal , Drug Delivery Systems , Humans , Immunity, Innate/immunology , Immunologic Factors/adverse effects , Immunologic Factors/immunology , Mice , Proteins/adverse effects , Proteins/immunology , T-Lymphocytes/immunology
2.
J Clin Pharmacol ; 57(3): 394-400, 2017 03.
Article in English | MEDLINE | ID: mdl-27557786

ABSTRACT

Nivolumab is a fully human IgG4 monoclonal antibody targeting the programmed death-1 (PD-1) receptor that blocks interactions between PD-1 and its ligands on tumor cells to prevent T-cell exhaustion in patients with cancer. It has demonstrated efficacy in multiple tumor types, including melanoma, non-small-cell lung cancer, and renal cell carcinoma. This analysis assessed the immunogenicity of nivolumab and its impact on pharmacokinetics, safety, and efficacy in patients with solid tumors enrolled in 6 clinical studies. The incidence and prevalence of antidrug antibodies (ADAs) were determined by validated electrochemiluminescence assays in samples collected during nivolumab treatment and up to 100 days after the last dose. Confirmed positive samples from the 6 studies were also tested for presence of neutralizing antibodies (NAbs). Among 1086 nivolumab-treated patients, 138 patients (12.7%) were ADA positive (relative to baseline), only 3 (0.3%) of whom were persistently positive for ADA, and 9 (0.8%) were NAb positive at 1 time point. The presence of ADAs was not associated with hypersensitivity, infusion reactions, or loss of efficacy and had minimal impact on nivolumab clearance. Additionally, the presence of NAbs was not associated with loss of efficacy. In conclusion, immunogenicity of nivolumab is not clinically meaningful.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Antibodies/immunology , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/immunology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Clinical Trials as Topic , Humans , Nivolumab
3.
Bioanalysis ; 8(1): 1-9, 2016.
Article in English | MEDLINE | ID: mdl-26648322

ABSTRACT

This article provides an overview of the information and factors relevant to designing bioanalytical strategies in support of in vivo nonclinical and clinical studies of protein therapeutics. The summarized information includes representative types of the therapeutic proteins, their key structural characteristics, the relationship between post-translational modifications and function, issues during purification and formulation, PK of therapeutic proteins and immunogenicity. The effect of each of those on bioanalysis strategy has been pointed out. The impacts of structural variant and 'free'/'bound' forms on PK assessment have been discussed.


Subject(s)
Chemistry Techniques, Analytical/methods , Proteins/analysis , Proteins/therapeutic use , Animals , Humans , Protein Processing, Post-Translational , Proteins/metabolism , Proteins/pharmacokinetics
4.
Anal Chem ; 85(20): 9859-67, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24024648

ABSTRACT

An LC-MS/MS assay was developed and fully validated for the simultaneous quantitation of two coadministered human monoclonal antibodies (mAbs), mAb-A and mAb-B of IgG4 subclass, in monkey serum. The total serum proteins were digested with trypsin at 50 °C for 30 min after methanol denaturation and precipitation, dithiothreitol reduction, and iodoacetamide alkylation. The tryptic peptides were chromatographically separated with a C18 column (2.1 × 100 mm, 1.7 µm) with mobile phases of 0.1% formic acid in water and acetonitrile. Four peptides, a unique peptide for each mAb and two confirmatory peptides from different antibody domains, were simultaneously quantified by LC-MS/MS in the multiple reaction-monitoring mode. Stable isotopically labeled peptides with flanking amino acids on C- and N-terminals were used as internal standards to minimize the variability during sample processing and detection. The LC-MS/MS assay showed lower limit of quantitation (LLOQ) at 5 µg/mL for mAb-A and 25 µg/mL for mAb-B. The intra- and interassay precision (%CV) was within 10.0% and 8.1%, respectively, and the accuracy (%Dev) was within ±5.4% for all the peptides. Other validation parameters, including sensitivity, selectivity, dilution linearity, processing recovery and matrix effect, autosampler carryover, run size, stability, and data reproducibility, were all evaluated. The confirmatory peptides played a critical role in confirming quantitation accuracy and the integrity of the drugs in the study samples. The robustness of the LC-MS/MS assay and the data agreement with the ligand binding data demonstrated that LC-MS/MS is a reliable and complementary approach for the quantitation of coadministered antibody drugs.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/blood , Blood Chemical Analysis/methods , Macaca fascicularis/blood , Tandem Mass Spectrometry , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Chemical Precipitation , Chromatography, Liquid , Feasibility Studies , Humans , Protein Denaturation , Time Factors , Trypsin/metabolism
5.
J Immunol ; 183(11): 7129-39, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19917701

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are key regulators of the innate immune response, yet their direct role as APCs in the adaptive immune response is unclear. We found that unlike conventional DCs, immune complex (IC) exposed murine pDCs neither up-regulated costimulatory molecules nor activated Ag-specific CD4(+) and CD8(+) T cells. The inability of murine pDCs to promote T cell activation was due to inefficient proteolytic processing of internalized ICs. This defect in the IC processing capacity of pDCs results from a lack of activating FcgammaR expression (FcgammaRI, III, IV) and the dominant expression of the inhibitory receptor FcgammaRIIB. Consistent with this idea, transgenic expression of the activating human FcgammaRIIA gene, not present in the mouse genome, recapitulated the human situation and rescued IC antigenic presentation capacity by murine pDCs. The selective expression of FcgammaRIIB by murine pDCs was not strain dependent and was maintained even following stimulation with TLR ligands and inflammatory cytokines. The unexpected difference between the mouse and human in the expression of activating/inhibitory FcgammaRs has implications for the role of pDCs in Ab-modulated autoimmunity and anti-viral immunity.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Receptors, IgG/biosynthesis , Animals , Antigen-Antibody Complex/immunology , Flow Cytometry , Humans , Mice , Mice, Knockout , Microscopy, Confocal , Receptors, IgG/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
6.
J Immunol ; 179(12): 8051-8, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18056345

ABSTRACT

Receptor for advanced glycation end products (RAGE) is an activation receptor triggered by inflammatory S100/calgranulins and high mobility group box-1 ligands. We have investigated the importance of RAGE on Ag priming of T cells in murine models in vivo. RAGE is inducibly up-regulated during T cell activation. Transfer of RAGE-deficient OT II T cells into OVA-immunized hosts resulted in reduced proliferative responses that were further diminished in RAGE-deficient recipients. Examination of RAGE-deficient dendritic cells did not reveal functional impairment in Ag presentation, maturation, or migratory capacities. However, RAGE-deficient T cells showed markedly impaired proliferative responses in vitro to nominal and alloantigens, in parallel with decreased production of IFN-gamma and IL-2. These data indicate that RAGE expressed on T cells is required for efficient priming of T cells and elucidate critical roles for RAGE engagement during cognate dendritic cell-T cell interactions.


Subject(s)
Antigen Presentation , Receptors, Immunologic/metabolism , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Cell Movement , Dendritic Cells/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Receptor for Advanced Glycation End Products , Receptors, Immunologic/analysis , Receptors, Immunologic/genetics , Th1 Cells/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
7.
J Immunol ; 178(10): 6217-26, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17475849

ABSTRACT

The uptake of immune complexes by FcRs on APCs augments humoral and cellular responses to exogenous Ag. In this study, CD11c+ dendritic cells are shown to be responsible in vivo for immune complex-triggered priming of T cells. We examine the consequence of Ab-mediated uptake of self Ag by dendritic cells in the rat insulin promoter-membrane OVA model and identify a role for the inhibitory FcgammaRIIB in the maintenance of peripheral CD8 T cell tolerance. Effector differentiation of diabetogenic OT-I CD8+ T cells is enhanced in rat insulin promoter-membrane OVA mice lacking FcgammaRIIB, resulting in a high incidence of diabetes. FcgammaRIIB-mediated inhibition of CD8 T cell priming results from suppression of both DC activation and cross-presentation through activating FcgammaRs. Further FcgammaRIIB on DCs inhibited the induction of OVA-specific Th1 effectors, limiting Th1-type differentiation and memory T cell accumulation. In these MHC II-restricted responses, the presence of FcgammaRIIB only modestly affected initial CD4 T cell proliferative responses, suggesting that FcgammaRIIB limited effector cell differentiation primarily by inhibiting DC activation. Thus, FcgammaRIIB can contribute to peripheral tolerance maintenance by inhibiting DC activation alone or by also limiting processing of exogenously acquired Ag.


Subject(s)
Antigens, CD/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Down-Regulation/immunology , Immune Tolerance , Receptors, IgG/physiology , Th1 Cells/immunology , Animals , Antigen-Antibody Complex/metabolism , Antigen-Antibody Complex/physiology , Antigens, CD/genetics , Autoantigens/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cross-Priming/genetics , Cross-Priming/immunology , Endocytosis/genetics , Endocytosis/immunology , Immune Tolerance/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/antagonists & inhibitors , Ovalbumin/biosynthesis , Ovalbumin/physiology , Receptors, IgG/deficiency , Receptors, IgG/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Th1 Cells/metabolism
8.
J Clin Invest ; 117(5): 1361-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17446931

ABSTRACT

We have developed a model of autoimmunity to investigate autoantibody-mediated cross-presentation of self antigen. RIP-mOVA mice, expressing OVA in pancreatic beta cells, develop severe autoimmune diabetes when given OT-I cells (OVA-specific CD8(+) T cells) and anti-OVA IgG but not when given T cells alone. Anti-OVA IgG is not directly injurious to the islets but rather enhances cross-presentation of apoptotic islet antigen to the OT-I cells, leading to their differentiation into potent effector cells. Antibody-driven effector T cell activation is dependent on the presence of activating Fc receptors for IgG (FcgammaRs) and cross-priming DCs. As a consequence, diabetes incidence and severity was reduced in mice lacking activating FcgammaRs. An intact complement pathway was also required for disease development, as C3 deficiency was also partially protective. C3-deficient animals exhibited augmented T cell priming overall, indicating a proinflammatory role for complement activation after the T cell priming phase. Thus, we show that autoreactive antibody can potently enhance the activation of effector T cells in response to cross-presented self antigen, thereby contributing to T cell-mediated autoimmunity.


Subject(s)
Autoantibodies/physiology , Autoantigens/immunology , Cross-Priming/immunology , Immune Tolerance , T-Lymphocytes/immunology , Animals , Autoantigens/metabolism , Cells, Cultured , Cross-Priming/genetics , Immune Tolerance/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/metabolism
9.
Immunity ; 23(5): 503-14, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16286018

ABSTRACT

Dendritic cells process internalized antigens to present degradative products on MHC for TCR recognition. Because antigen-exposed DCs also induce humoral immunity, DCs must also retain antigen in its native state for the engagement of BCR on B cells. Here, we demonstrate that antigen endocytosed by the inhibitory Fc receptor, FcgammaRIIB, accesses a non-degradative intracellular vesicular compartment that recycles to the cell surface, enabling interaction of native antigen with BCR on B cells. Immunization with IgG-opsonized, T independent antigens leads to enhanced humoral responses in a FcgammaRIIB and complement dependent manner. IC-loaded DCs trafficking to the splenic marginal zone can prime a T independent response in an FcgammaRIIB-dependent manner. Thus dendritic cells are equipped with both non-degradative and degradative antigen uptake pathways to facilitate antigen presentation to both B and T cells.


Subject(s)
Antigens/immunology , Antigens/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis , Animals , Antigen Presentation , Antigens, CD/immunology , Cell Proliferation , Cells, Cultured , Dendritic Cells/cytology , Lymphocyte Activation , Mice , Mice, Knockout , Receptors, IgG/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...