Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 20(2): 357-366, 2021 02.
Article in English | MEDLINE | ID: mdl-33298591

ABSTRACT

Targeted T-cell redirection is a promising field in cancer immunotherapy. T-cell bispecific antibodies (TCB) are novel antibody constructs capable of binding simultaneously to T cells and tumor cells, allowing cross-linking and the formation of immunologic synapses. This in turn results in T-cell activation, expansion, and tumor killing. TCB activity depends on system-related properties such as tumor target antigen expression as well as antibody properties such as binding affinities to target and T cells. Here, we developed a systems model integrating in vitro data to elucidate further the mechanism of action and to quantify the cytotoxic effects as the relationship between targeted antigen expression and corresponding TCB activity. In the proposed model, we capture relevant processes, linking immune synapse formation to T-cell activation, expansion, and tumor killing for TCBs in vitro to differentiate the effect between tumor cells expressing high or low levels of the tumor antigen. We used cibisatamab, a TCB binding to carcinoembryonic antigen (CEA), to target different tumor cell lines with high and low CEA expression in vitro We developed a model to capture and predict our observations, as a learn-and-confirm cycle. Although full tumor killing and substantial T-cell activation was observed in high expressing tumor cells, the model correctly predicted partial tumor killing and minimal T-cell activation in low expressing tumor cells when exposed to cibisatamab. Furthermore, the model successfully predicted cytotoxicity across a wide range of tumor cell lines, spanning from very low to high CEA expression.


Subject(s)
Antibodies, Bispecific/metabolism , T-Lymphocytes/metabolism , Animals , Cell Line, Tumor , Humans
2.
Sci Transl Med ; 12(534)2020 03 11.
Article in English | MEDLINE | ID: mdl-32161104

ABSTRACT

PD-L1/PD-1 blocking antibodies have demonstrated therapeutic efficacy across a range of human cancers. Extending this benefit to a greater number of patients, however, will require a better understanding of how these therapies instigate anticancer immunity. Although the PD-L1/PD-1 axis is typically associated with T cell function, we demonstrate here that dendritic cells (DCs) are an important target of PD-L1 blocking antibody. PD-L1 binds two receptors, PD-1 and B7.1 (CD80). PD-L1 is expressed much more abundantly than B7.1 on peripheral and tumor-associated DCs in patients with cancer. Blocking PD-L1 on DCs relieves B7.1 sequestration in cis by PD-L1, which allows the B7.1/CD28 interaction to enhance T cell priming. In line with this, in patients with renal cell carcinoma or non-small cell lung cancer treated with atezolizumab (PD-L1 blockade), a DC gene signature is strongly associated with improved overall survival. These data suggest that PD-L1 blockade reinvigorates DC function to generate potent anticancer T cell immunity.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung , Immunotherapy , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Dendritic Cells , Humans , Lung Neoplasms/drug therapy
3.
MAbs ; 6(6): 1608-20, 2014.
Article in English | MEDLINE | ID: mdl-25484038

ABSTRACT

Anti-cytokine autoantibodies have been widely reported to be present in human plasma, both in healthy subjects and in patients with underlying autoimmune conditions, such as autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED) or thymic epithelial neoplasms. While often asymptomatic, they can cause or facilitate a wide range of diseases including opportunistic infections. The potential therapeutic value of specific neutralizing anti-cytokine autoantibodies has not been thoroughly investigated. Here we used mammalian cell display to isolate IL17A-specific antibodies from a thymoma patient with proven high-titer autoantibodies against the same. We identified 3 distinct clonotypes that efficiently neutralized IL17A in a cell-based in vitro assay. Their potencies were comparable to those of known neutralizing antibodies, including 2, AIN457 (secukinumab) and ixekizumab that are currently in clinical development for the treatment of various inflammatory disorders. These data clearly demonstrate that the human autoantibody repertoire can be mined for antibodies with high therapeutic potential for clinical development.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Autoantibodies/immunology , Interleukin-17/immunology , Thymoma/immunology , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/isolation & purification , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/isolation & purification , Antibody Affinity/immunology , Autoantibodies/genetics , Autoantibodies/isolation & purification , Cell Line , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Interleukin-17/genetics , Molecular Sequence Data , Neutralization Tests , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Proteins/immunology , Sequence Homology, Amino Acid , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Thymoma/blood
4.
J Allergy Clin Immunol ; 126(2): 375-83, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20624641

ABSTRACT

BACKGROUND: Allergen-specific desensitization (SIT) is the most effective therapy for allergies. Although allergen-specific antibodies have an important role in the process, mechanisms of IgG-mediated inhibition of allergic reactions are not well defined. OBJECTIVE: We investigated mechanisms by which SIT-induced allergen-specific IgGs inhibit allergic reactions. METHODS: We generated mAbs that recognize 3 nonoverlapping epitopes of the major cat allergen Fel d 1. Each of the mAbs was produced as an IgE and different IgG isotype. RESULTS: IgEs against 2 nonoverlapping epitopes on Fel d 1 are necessary and sufficient to sensitize mast cells for maximal FcepsilonRI signaling and degranulation on exposure to monomeric Fel d 1. IgE antibodies of a third specificity did not further increase mast cell degranulation, indicating that formation of large FcepsilonRI clusters are not required to induce maximal activation of mast cells. A single IgG that was specific for an epitope different from those recognized by the IgEs was a potent inhibitor of Fel d 1-mediated mast cell activation in vitro and in vivo. This inhibition required Fcgamma receptor-IIB. In human beings, IgGs of a single specificity were able to block degranulation of basophils from individuals with cat allergy. The inhibitory potential of these antibodies increased when larger allergen-IgG complexes were formed. CONCLUSIONS: These data reconcile conflicting theories in the literature and might explain the reason IgE levels do not necessarily decrease during therapy, despite clinical efficacy. These findings have important implications for vaccine design.


Subject(s)
Allergens/immunology , Antibodies, Monoclonal/pharmacology , Desensitization, Immunologic , Glycoproteins/immunology , Hypersensitivity/therapy , Immunoglobulin G/pharmacology , Allergens/pharmacology , Animals , Antibodies, Monoclonal/immunology , Epitopes/immunology , Glycoproteins/antagonists & inhibitors , Glycoproteins/pharmacology , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, IgE/genetics , Receptors, IgE/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Vaccines/genetics , Vaccines/immunology
5.
Virol J ; 6: 224, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-20025741

ABSTRACT

Influenza virus infection is a prevalent disease in humans. Antibodies against hemagglutinin have been shown to prevent infection and hence hemagglutinin is the major constituent of current vaccines. Antibodies directed against the highly conserved extracellular domain of M2 have also been shown to mediate protection against Influenza A infection in various animal models. Active vaccination is generally considered the best approach to combat viral diseases. However, passive immunization is an attractive alternative, particularly in acutely exposed or immune compromized individuals, young children and the elderly. We recently described a novel method for the rapid isolation of natural human antibodies by mammalian cell display. Here we used this approach to isolate human monoclonal antibodies directed against the highly conserved extracellular domain of the Influenza A M2 protein. The identified antibodies bound M2 peptide with high affinities, recognized native cell-surface expressed M2 and protected mice from a lethal influenza virus challenge. Moreover, therapeutic treatment up to 2 days after infection was effective, suggesting that M2-specific monoclonals have a great potential as immunotherapeutic agents against Influenza infection.


Subject(s)
Antibodies, Monoclonal , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae Infections , Viral Matrix Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Cell Line , Cricetinae , Disease Models, Animal , Female , Humans , Immunization, Passive , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Treatment Outcome , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 106(28): 11673-8, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19564598

ABSTRACT

Suppression by natural CD4(+)CD25(+) regulatory T cells (Tregs) is one mechanism by which tolerance is maintained. However, the way in which Tregs mediate suppression is not well understood. Here, we show that secreted phospholipase A2 (sPLA2)-IID is selectively produced by Tregs. sPLA2-IID is a potent mediator of Treg function, because it strongly suppressed proliferation of CD4(+) and CD8(+) T cells in vitro and in vivo in a manner independent of its catalytic activity. Furthermore, sPLA2-IID promoted the differentiation of Tregs, presumably via attenuating signaling through the PI3K/Akt/mammalian target of rapamycin pathway. Importantly, administration of a sPLA2-IID-Fc fusion protein inhibited disease development in murine models of colitis and multiple sclerosis, suggesting that sPLA2-IID's immunosuppressive function might be exploited therapeutically.


Subject(s)
Cell Differentiation/immunology , Cell Proliferation/drug effects , Colitis/prevention & control , Group II Phospholipases A2/pharmacology , Multiple Sclerosis/prevention & control , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Animals , Cell Differentiation/drug effects , Colitis/immunology , DNA Primers/genetics , Flow Cytometry , Group II Phospholipases A2/metabolism , Mice , Multiple Sclerosis/immunology , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
7.
Proc Natl Acad Sci U S A ; 105(38): 14336-41, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18812621

ABSTRACT

Due to their low immunogenicity in patients, humanized or fully human mAbs are becoming increasingly important for the treatment of a growing number of diseases, including cancer, infections, and immune disorders. Here, we describe a technology allowing for the rapid isolation of fully human mAbs. In contrast to previously described methods, B cells specific for an antigen of interest are directly isolated from peripheral blood mononuclear cells (PBMC) of human donors. Recombinant, antigen-specific single-chain Fv (scFv) libraries are generated from this pool of B cells and screened by mammalian cell surface display by using a Sindbis virus expression system. This method allows isolating antigen-specific antibodies by a single round of FACS. The variable regions (VRs) of the heavy chains (HCs) and light chains (LCs) are isolated from positive clones and recombinant fully human antibodies produced as whole IgG or Fab fragments. In this manner, several hypermutated high-affinity antibodies binding the Qbeta virus like particle (VLP), a model viral antigen, as well as antibodies specific for nicotine were isolated. All antibodies showed high expression levels in cell culture. The human nicotine-specific mAbs were validated preclinically in a mouse model. Thus, the technology presented here allows for rapid isolation of high-affinity, fully human antibodies with therapeutic potential from human volunteers.


Subject(s)
Allolevivirus/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Mammals , Peptide Library , Animals , Antibodies, Monoclonal/genetics , Antibodies, Viral/genetics , Brain/metabolism , Capsid Proteins/immunology , Flow Cytometry , Humans , Immunization, Passive , Leukocytes, Mononuclear/immunology , Mice , Nicotine/immunology , Sindbis Virus/genetics , Sindbis Virus/metabolism
8.
Mol Immunol ; 45(10): 2727-33, 2008 May.
Article in English | MEDLINE | ID: mdl-18372042

ABSTRACT

Plasma cells are the main producers of antibody and key effector cells of the immune system. Despite their importance, analytics of plasma cells still suffers from the limited availability of specific markers. Currently, plasma cell identification relies on the expression of a single marker, CD138/syndecan-1. However, syndecan-1 is widely expressed on various cell types outside the hematopoietic compartment, and furthermore, not expressed on all subsets of plasma cells. To discover novel surface markers, a differential screening followed by signal sequence trap cloning was developed, leading to the identification of mouse Ly-6K (mLy-6K). Expression profiling confirmed that mLy-6K is expressed by plasma cells but not B cells or tissues not containing plasma cells. Expression at the surface of plasma cells isolated from spleen, lymph node, bone marrow, and lamina propria of the small intestine was demonstrated at the protein level using a polyclonal rabbit antibody. This novel plasma cell marker shows promise to help broaden our understanding of plasma cell differentiation and function.


Subject(s)
Antigens, Ly/metabolism , Plasma Cells/metabolism , Amino Acid Sequence , Animals , Antigens, Ly/chemistry , Antigens, Ly/genetics , Biomarkers/chemistry , Biomarkers/metabolism , Blotting, Northern , Cell Line , Cell Membrane/metabolism , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Plasma Cells/cytology , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...