Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Oncol ; 14: 1346502, 2024.
Article in English | MEDLINE | ID: mdl-38577337

ABSTRACT

Introduction: Although checkpoint inhibitors (CPIs) have improved outcomes for patients with metastatic melanoma, those progressing on CPIs have limited therapeutic options. To address this unmet need and overcome CPI resistance mechanisms, novel immunotherapies, such as T-cell engaging agents, are being developed. The use of these agents has sometimes been limited by the immune response mounted against them in the form of anti-drug antibodies (ADAs), which is challenging to predict preclinically and can lead to neutralization of the drug and loss of efficacy. Methods: TYRP1-TCB (RO7293583; RG6232) is a T-cell engaging bispecific (TCB) antibody that targets tyrosinase-related protein 1 (TYRP1), which is expressed in many melanomas, thereby directing T cells to kill TYRP1-expressing tumor cells. Preclinical studies show TYRP1-TCB to have potent anti-tumor activity. This first-in-human (FIH) phase 1 dose-escalation study characterized the safety, tolerability, maximum tolerated dose/optimal biological dose, and pharmacokinetics (PK) of TYRP1-TCB in patients with metastatic melanoma (NCT04551352). Results: Twenty participants with cutaneous, uveal, or mucosal TYRP1-positive melanoma received TYRP1-TCB in escalating doses (0.045 to 0.4 mg). All participants experienced ≥1 treatment-related adverse event (TRAE); two participants experienced grade 3 TRAEs. The most common toxicities were grade 1-2 cytokine release syndrome (CRS) and rash. Fractionated dosing mitigated CRS and was associated with lower levels of interleukin-6 and tumor necrosis factor-alpha. Measurement of active drug (dual TYPR1- and CD3-binding) PK rapidly identified loss of active drug exposure in all participants treated with 0.4 mg in a flat dosing schedule for ≥3 cycles. Loss of exposure was associated with development of ADAs towards both the TYRP1 and CD3 domains. A total drug PK assay, measuring free and ADA-bound forms, demonstrated that TYRP1-TCB-ADA immune complexes were present in participant samples, but showed no drug activity in vitro. Discussion: This study provides important insights into how the use of active drug PK assays, coupled with mechanistic follow-up, can inform and enable ongoing benefit/risk assessment for individuals participating in FIH dose-escalation trials. Translational studies that lead to a better understanding of the underlying biology of cognate T- and B-cell interactions, ultimately resulting in ADA development to novel biotherapeutics, are needed.

2.
MAbs ; 14(1): 2143009, 2022.
Article in English | MEDLINE | ID: mdl-36394299

ABSTRACT

ABBREVIATIONS: ADA Anti-Drug Antibodies; BCR B Cell Receptor; BId Idiotype-specific B Cell; BiTE Bispecific T cell Engager; BMC Bone Marrow Chimeric Mice; BSA Bovine Serum Albumin; CDR Complementary Determining Region; CEA Carcinoembryonic Antigen; CIT Cancer Immunotherapy; CitAbs Cancer Immunotherapy Antibodies; DC Dendritic Cell; ELISA Enzyme-Linked Immunosorbent Assay; FcRn Neonatal Fc Receptor; FcyR Fc gamma Receptor; GM-CSF Granulocyte-Macrophage Colony Stimulating Factor; gMFI Geometric Mean Fluorescence Intensity; H Heavy Chain; IC Immune Complex; Id Idiotype; IgA Immunoglobulin alpha; IgG1 Immunoglobulin gamma 1; IL-2 Interleukin 2; IL-2R Interleukin 2 Receptor; IL2v Interleukin 2 Variant; IVIG1 Intravenous Immunoglobulin 1; KLH Keyhole Limpet Hemocyanin; L Light Chain; MAPPs MHC-associated Peptide Proteomics; MHC Major Histocompatibility Complex; PBMC Peripheral Blood Mononuclear Cells; PBS Phosphate Buffered Saline; SHM Somatic Hypermutation; scFv Single-chain Variable Fragment; TCR T cell Receptor; TFc Fc-specific T cell; TId Id-specific T cell; UV Ultraviolet; V Variable.


Subject(s)
Immunoglobulin G , Neoplasms , Humans , Mice , Animals , Interleukin-2 , Mice, Transgenic , Leukocytes, Mononuclear , Immunotherapy
3.
MAbs ; 14(1): 1993522, 2022.
Article in English | MEDLINE | ID: mdl-34923896

ABSTRACT

A major impediment to successful use of therapeutic protein drugs is their ability to induce anti-drug antibodies (ADA) that can alter treatment efficacy and safety in a significant number of patients. To this aim, in silico, in vitro, and in vivo tools have been developed to assess sequence and other liabilities contributing to ADA development at different stages of the immune response. However, variability exists between similar assays developed by different investigators due to the complexity of assays, a degree of uncertainty about the underlying science, and their intended use. The impact of protocol variations on the outcome of the assays, i.e., on the immunogenicity risk assigned to a given drug candidate, cannot always be precisely assessed. Here, the Non-Clinical Immunogenicity Risk Assessment working group of the European Immunogenicity Platform (EIP) reviews currently used assays and protocols and discusses feasibility and next steps toward harmonization and standardization.


Subject(s)
Antibodies, Monoclonal , Immunoconjugates , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Drug Evaluation, Preclinical , Humans , Immunoconjugates/adverse effects , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Risk Assessment
4.
J Pharm Sci ; 110(7): 2575-2584, 2021 07.
Article in English | MEDLINE | ID: mdl-33812888

ABSTRACT

Biotherapeutics have revolutionized our ability to treat life-threatening diseases. Despite clinical success, the use of biotherapeutics has sometimes been limited by the immune response mounted against them in the form of anti-drug antibodies (ADAs). The multifactorial nature of immunogenicity has prevented a standardized approach for assessing this and each of the assessment methods developed so far does not exhibit high enough reliability to be used alone, due to limited predictiveness. This prompted the Roche Pharma Research and Early Development (pRED) Immunogenicity Working Group to establish an internal preclinical immunogenicity toolbox of in vitro/in vivo approaches and accompanying guidelines for a harmonized assessment and management of immunogenicity in early development. In this article, the complex factors influencing immunogenicity and their associated clinical ramifications are discussed to highlight the importance of an end-to-end approach conducted from lead optimization to clinical candidate selection. We then examine the impact of the resulting lead candidate categorization on the design and implementation of a multi-tiered ADA/immunogenicity assay strategy prior to phase I (entry into human) through early clinical development. Ultimately, the Immunogenicity Toolbox ensures that Roche pRED teams are equipped to address immunogenicity in a standardized manner, paving the way for lifesaving products with improved safety and efficacy.


Subject(s)
Antibodies , Immunologic Factors , Humans , Immunotherapy , Reproducibility of Results
5.
Mol Pharm ; 14(4): 1292-1299, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28206769

ABSTRACT

A current concern with the use of therapeutic proteins is the likely presence of aggregates and submicrometer, subvisible, and visible particles. It has been proposed that aggregates and particles may lead to unwanted increases in the immune response with a possible impact on safety or efficacy. The aim of this study was thus to evaluate the ability of subvisible particles of a therapeutic antibody to break immune tolerance in an IgG1 transgenic mouse model and to understand the particle attributes that might play a role in this process. We investigated the immunogenic properties of subvisible particles (unfractionated, mixed populations, and well-defined particle size fractions) using a transgenic mouse model expressing a mini-repertoire of human IgG1 (hIgG1 tg). Immunization with proteinaceous subvisible particles generated by artificial stress conditions demonstrated that only subvisible particles bearing very extensive chemical modifications within the primary amino acid structure could break immune tolerance in the hIgG1 transgenic mouse model. Protein particles exhibiting low levels of chemical modification were not immunogenic in this model.


Subject(s)
Immune Tolerance/immunology , Immunoglobulin G/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibody Formation/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Particle Size
6.
AAPS J ; 18(2): 354-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26603888

ABSTRACT

The purpose of this study is to test the feasibility of neonatal immune tolerance induction in mice to enable long-term pharmacokinetic studies with immunogenic therapeutic monoclonal antibodies (mAb). Neonatal immune tolerance was induced by transfer of a mAb to neonatal mice via colostrum from nursing mother mice treated with two subcutaneous doses of a tolerogen starting within the first 24 h after delivery. Adalimumab and efalizumab were administered as tolerogens at various dose levels. Tolerance induction was evaluated in the offspring after reaching adulthood at 8 weeks of age. After a single intravenous injection of the same mAb as used for tolerance induction, the pharmacokinetics of the mAb and formation of anti-drug antibodies (ADA) in plasma were assessed using ELISA. Tolerance induction to adalimumab was achieved in a maternal dose-dependent manner. Adalimumab immune-tolerant offspring showed a slower adalimumab clearance (4.24 ± 0.32 mL/day/kg) as compared to the control group (12.09 ± 3.81 mL/day/kg). In the control group, accelerated clearance started 7 days after adalimumab dosing, whereas immune-tolerant offspring showed a log-linear terminal concentration-time course. In the offspring, the absence of predose ADA levels was indicative of successful tolerance induction. The second test compound efalizumab was not immunogenic in mice under our experimental conditions. Overall, the present study demonstrated the suitability of neonatal immune tolerance induction for a 4-week single dose study in adult mice with a human therapeutic mAb that is otherwise immunogenic in laboratory animals.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Formation/drug effects , Antibody Formation/immunology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Adalimumab/pharmacology , Animals , Animals, Newborn , Antibodies, Monoclonal/blood , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology , Time Factors
7.
Pharm Res ; 32(7): 2344-59, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25630815

ABSTRACT

PURPOSE: Protein aggregates have been discussed as a potential risk factor related to immunogenicity. Here we developed a novel human IgG transgenic (tg) mouse system expressing a mini-repertoire of human IgG1 antibodies (Abs) for the assessment of immunogenic properties of human mAb preparations. METHODS: Transgenic mice were generated using germline versions of the human Ig heavy chain γ1 (IgH-γ1), and the human Ig light chain (IgL) κ and λ genes. Only the soluble form of human IgH-γ1 was used to avoid expression of the membrane Ig-H chain and concomitant allelic exclusion of endogenous murine Ig genes. IgG1 aggregates were generated by different stress conditions such as process-related, low pH and exposure to artificial light. RESULTS: The expression of human Ig proteins induced immunological tolerance to a broad range of human IgG1 molecules in the tg mice. Immunization with IgG1 aggregates demonstrated that soluble oligomers induced by significant light-exposure and carrying neo-epitopes induced a strong immune response in tg mice. In contrast, Ab aggregates alone and monomers with neo-epitopes were not immunogenic. CONCLUSION: This mouse model is able to recognize immunogenic modifications of human IgG1. While the degree of stress-induced aggregation varies for different mAbs, our findings using a particular mAb (mAb1) demonstrate that non-covalently modified aggregates do not break tolerance, contrary to widely held opinion. The immunogenic potential of soluble aggregates of human IgG strongly depends on the presence of neo-epitopes resulting from harsh stress conditions, i.e. extensive exposure to artificial light.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Mice, Transgenic/immunology , Protein Aggregates/immunology , Animals , Antibodies, Monoclonal/genetics , Antibody Formation , Base Sequence , Flow Cytometry , Humans , Immune Tolerance , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Mice, Transgenic/genetics , Molecular Sequence Data , Protein Aggregates/genetics , Stress, Psychological/immunology , Transgenes
8.
J Immunol ; 192(12): 5499-508, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24821969

ABSTRACT

Extensive studies have been undertaken to describe naive B cells differentiating into memory B cells at a cellular and molecular level. However, relatively little is known about the fate of memory B cells upon Ag re-encounter. We have previously established a system based on virus-like particles (VLPs), which allows tracking of VLP-specific B cells by flow cytometry as well as histology. Using allotype markers, it is possible to adoptively transfer memory B cells into a naive mouse and track responses of naive and memory B cells in the same mouse under physiological conditions. We have observed that VLP-specific memory B cells quickly differentiated into plasma cells that drove the early onset of a strong humoral IgG response. However, neither IgM(+) nor IgG(+) memory B cells proliferated extensively or entered germinal centers. Remarkably, plasma cells derived from memory B cells preferentially homed to the bone marrow earlier and secreted increased levels of Abs when compared with primary plasma cells derived from naive B cells. Hence, memory B cells have the unique phenotype to differentiate into highly effective secondary plasma cells.


Subject(s)
Allolevivirus/immunology , Antibodies, Viral/immunology , Cell Differentiation/immunology , Immunologic Memory , Plasma Cells/immunology , Animals , Immunization , Mice
9.
J Autoimmun ; 55: 33-41, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24786898

ABSTRACT

Non-resolving inflammation is a major contributor to chronic disease pathogenesis, including that of cancer, chronic obstructive pulmonary disease, asthma, arthritis, inflammatory bowel disease, multiple sclerosis and obesity. Some cytokines, such as IL-1α and IL-33, may act as endogenous alarmins that contribute to non-resolving inflammation. These cytokines are constitutively expressed in the nucleus and are thought to promote inflammation only upon release during tissue damage or cell necrosis. However, the importance of their nuclear localization in immune homeostasis is not fully understood. We describe herein a novel mouse model in which the nuclear localization signal of IL-33 is abolished and demonstrate for the first time that, alone, altered subcellular localization of IL-33 dramatically affects immune homeostasis. Heterozygous IL33(tm1/+) mice display elevated serum IL-33 levels, indicating that IL-33 is constitutively released when not actively targeted to the nucleus. IL33(tm1/+) mice succumb to lethal inflammation characterized by eosinophil-dominated immune cell infiltration of multiple organs. The profound inflammatory phenotype is dependent on mediators downstream of ST2 as ST2-null mice are protected in spite of high serum IL-33 levels. Importantly, IL-33 transcript levels in this knock-in mouse model remain under endogenous control. We adopt the term "nuclear alarmin" to describe a danger signal that is primarily regulated by nuclear compartmentalization in this fashion.


Subject(s)
Cell Nucleus/immunology , Homeostasis/immunology , Interleukins/immunology , Nuclear Localization Signals/immunology , Receptors, Interleukin/immunology , Animals , Cell Nucleus/genetics , Cell Nucleus/pathology , Homeostasis/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Localization Signals/genetics , Receptors, Interleukin/genetics
10.
Proc Natl Acad Sci U S A ; 109(50): 20566-71, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23169669

ABSTRACT

The lung is an important entry site for pathogens; its exposure to antigens results in systemic as well as local IgA and IgG antibodies. Here we show that intranasal administration of virus-like particles (VLPs) results in splenic B-cell responses with strong local germinal-center formation. Surprisingly, VLPs were not transported from the lung to the spleen in a free form but by B cells. The interaction between VLPs and B cells was initiated in the lung and occurred independently of complement receptor 2 and Fcγ receptors, but was dependent upon B-cell receptors. Thus, B cells passing through the lungs bind VLPs via their B-cell receptors and deliver them to local B cells within the splenic B-cell follicle. This process is fundamentally different from delivery of blood or lymph borne particulate antigens, which are transported into B cell follicles by binding to complement receptors on B cells.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/virology , Vaccines, Virus-Like Particle/immunology , Administration, Intranasal , Adoptive Transfer , Animals , Antibodies, Viral/biosynthesis , Antigens, Viral/administration & dosage , Antigens, Viral/blood , Cell Movement/immunology , Female , Immunoglobulin G/biosynthesis , Lung/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Receptors, Antigen, B-Cell/immunology , Receptors, Complement 3d/immunology , Receptors, IgG/immunology , Spleen/immunology , Spleen/virology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/blood
11.
Immunol Invest ; 39(4-5): 407-28, 2010.
Article in English | MEDLINE | ID: mdl-20450285

ABSTRACT

Immunoglobulin A (IgA) represents the primary line of protection against incoming pathogens since it is the predominant isotype on mucosal surfaces. Mucosal surfaces are constantly exposed to inhaled, digested and sexually transmitted agents and therefore highly susceptible to infection by invading pathogens. Such pathogens typically carry pathogen-associated molecular patterns (PAMPs) which primarily signal through Toll-like receptors (TLRs). TLRs belong to a family of pattern-recognition receptors that link the innate and the acquired immune system. TLR stimulation in professional antigen-presenting cells (APCs) such as dendritic cells (DCs) is crucial for an optimal cellular and humoral immune response to be induced. Moreover TLRs have been shown to improve humoral responses by direct stimulation of B cells. Herein we review recent data, which points to a pivotal role of TLR signalling in controlling T-cell dependent and independent IgA responses both at mucosal and systemic levels. A better understanding of these mechanisms may facilitate the use of TLR agonists as adjuvants and consequently improve the development of effective mucosal vaccines.


Subject(s)
Immunoglobulin A/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Toll-Like Receptors/immunology , Animals , Humans , Immunity, Mucosal , Immunoglobulin Class Switching , Infections/immunology , T-Lymphocytes/metabolism , Toll-Like Receptors/metabolism
12.
J Immunol ; 184(9): 4615-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20368279

ABSTRACT

IL-21 produced by follicular Th (Tfh) cells is an important regulator of Tfh cell development and B cell responses, including germinal center (GC) formation. However, whether defective GC formation and Ab responses are a consequence of impaired Tfh cells development or a B cell-intrinsic defect in IL-21-deficient mice requires clarification. To address this question, we generated chimeric mice lacking IL-21R exclusively on B cells. In this study, we demonstrate that GC reaction and B cell responses induced by immunization with virus-like particles were strongly reduced in both global and B cell-specific IL-21R-deficient mice. Interestingly, the presence of TLR7 ligand within virus-like particles largely restored defective GC reaction and Ab responses in global as well as in B cell-specific IL-21R-deficient mice. Hence, IL-21 acts directly on B cells and cooperates with TLR signaling for optimal B cell responses.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Interleukins/physiology , Membrane Glycoproteins/physiology , Signal Transduction/immunology , Toll-Like Receptor 7/physiology , Toll-Like Receptor 8/physiology , Allolevivirus/genetics , Allolevivirus/immunology , Animals , Antibodies, Viral/biosynthesis , B-Lymphocyte Subsets/virology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Gene Expression Regulation, Viral/immunology , Gene Products, env/biosynthesis , Gene Products, env/genetics , Gene Products, env/physiology , Germinal Center/cytology , Germinal Center/virology , Immunity, Innate/genetics , Immunoglobulin G/biosynthesis , Interleukins/metabolism , Ligands , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-21/deficiency , Receptors, Interleukin-21/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/virology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Virion/genetics , Virion/immunology
13.
J Immunol ; 183(6): 3788-99, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19710454

ABSTRACT

The mechanisms regulating systemic and mucosal IgA responses in the respiratory tract are incompletely understood. Using virus-like particles loaded with single-stranded RNA as a ligand for TLR7, we found that systemic vs mucosal IgA responses in mice were differently regulated. Systemic IgA responses following s.c. immunization were T cell independent and did not require TACI or TGFbeta, whereas mucosal IgA production was dependent on Th cells, TACI, and TGFbeta. Strikingly, both responses required TLR7 signaling, but systemic IgA depended upon TLR7 signaling directly to B cells whereas mucosal IgA required TLR7 signaling to lung dendritic cells and alveolar macrophages. Our data show that IgA switching is controlled differently according to the cell type receiving TLR signals. This knowledge should facilitate the development of IgA-inducing vaccines.


Subject(s)
Dendritic Cells/immunology , Immunoglobulin A/biosynthesis , Lung/immunology , Macrophages, Alveolar/immunology , Mucous Membrane/immunology , RNA/immunology , Animals , Membrane Glycoproteins , Mice , Mice, Knockout , T-Lymphocytes, Helper-Inducer , Toll-Like Receptor 7 , Transforming Growth Factor beta , Transmembrane Activator and CAML Interactor Protein
14.
Eur J Immunol ; 38(1): 114-26, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18081037

ABSTRACT

Intranasal (i.n.) immunization aims to induce local as well as systemic immune responses. In the present study, we assessed a vaccine platform based on virus-like particles (VLP) derived from the RNA phage Qbeta for i.n. immunization. We found that both i.n. and subcutaneous (s.c.) administration of Qbeta-VLP elicited strong and comparable specific IgG responses in serum and lung. Surprisingly, both routes also induced high levels of specific IgA in serum. In contrast, only i.n. administration of Qbeta-VLP resulted in local IgA production in the lung. Efficient induction of B cell responses by i.n. administration of VLP was further supported by the presence of large numbers of germinal centers (GC) as well as memory B cells in the spleen and plasma cells in the bone marrow. Results obtained for the VLP itself could be extended to an antigen covalently attached to it. Specifically, i.n. immunization of mice with VLP displaying the influenza virus derived ectodomain of the M2 protein resulted in strong M2-specific antibody responses as well as anti-viral protection. In contrast, i.n. immunization with VLP displaying p33 peptide, the major CTL epitope of lymphocytic choriomeningitis virus, induced relatively inefficient cytotoxic T cell responses, resulting in low numbers of specific T cells and poor effector cell differentiation. Taken together, these results suggest that effective antibody-based vaccines are achievable by i.n. administration of Qbeta-VLP displaying specific antigens.


Subject(s)
Administration, Intranasal , Immunity, Mucosal , RNA, Viral/administration & dosage , Vaccination/methods , Viral Vaccines/administration & dosage , Virion/immunology , Animals , Antibodies, Viral/blood , Antibody Formation , B-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Germinal Center/immunology , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , RNA, Viral/immunology , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Matrix Proteins/immunology , Viral Vaccines/immunology
15.
J Immunol ; 178(4): 2415-20, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277148

ABSTRACT

Although IgG2a is the most potent Ab isotype in the host response to viral and bacterial infections, the regulation of class switch recombination to IgG2a in vivo is not yet well understood. Recognition of pathogen-associated molecular patterns by dendritic cells expressing TLRs, like TLR7, recognizing ssRNA, or TLR9, recognizing DNA rich in nonmethylated CG motifs (CpG), favors induction of Th1 responses. It is generally assumed that these Th1 responses are responsible for the TLR-mediated induction of IgG2a. Using virus-like particles loaded with CpGs, we show here that TLR9 ligands can directly stimulate B cells to undergo isotype switching to IgG2a. Unexpectedly, TLR9 expression in non-B cells did not affect isotype switching in the Ab response against virus-like particles. Thus, TLR9 can regulate isotype switching to IgG2a directly by interacting with B cells rather than indirectly by inducing Th1 responses.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Toll-Like Receptor 9/immunology , Animals , B-Lymphocytes/cytology , Bacterial Infections/immunology , CpG Islands/immunology , Ligands , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , RNA/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Toll-Like Receptor 7/immunology , Virus Diseases/immunology
16.
J Immunol ; 178(1): 67-76, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17182541

ABSTRACT

Protective Ab levels can be maintained for years upon infection or vaccination. In this study, we studied the duration of Ab responses as a function of the life span of plasma cells and tested the role of persisting Ag in maintaining B cell memory. Our analysis of B cell responses induced in mice immunized with virus-like particles demonstrates the following: 1) Ab titers are long-lived, but decline continuously with a t(1/2) of approximately 80 days, which corresponds to the life span of plasma cells; 2) the germinal center (GC) reaction, which lasts for up to 100 days, is dependent on Ag associated with follicular dendritic cells; and 3) early GCs produce massive numbers of plasma and memory B cell precursors, whereas the late Ag-dependent GCs are dispensable for the maintenance of Ab levels and B cell memory.


Subject(s)
Antibodies/blood , Antigens/immunology , Immunologic Memory , Plasma Cells/immunology , Allolevivirus/immunology , Animals , Antigens/blood , Cell Proliferation , Dendritic Cells, Follicular/drug effects , Dendritic Cells, Follicular/immunology , Germinal Center/cytology , Germinal Center/immunology , Immunization , Immunoglobulins/pharmacology , Immunosuppression Therapy , Kinetics , Lymphotoxin beta Receptor/immunology , Mice , Mice, Inbred C57BL , Plasma Cells/cytology , Virion/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...