Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Clin Exp Med ; 24(1): 122, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38856863

ABSTRACT

Regulatory T cells (Tregs) are known to facilitate tumor progression by suppressing CD8+ T cells within the tumor microenvironment (TME), thereby also hampering the effectiveness of immune checkpoint inhibitors (ICIs). While systemic depletion of Tregs can enhance antitumor immunity, it also triggers undesirable autoimmune responses. Therefore, there is a need for therapeutic agents that selectively target Tregs within the TME without affecting systemic Tregs. In this study, as shown also by others, the chemokine (C-C motif) receptor 8 (CCR8) was found to be predominantly expressed on Tregs within the TME of both humans and mice, representing a unique target for selective depletion of tumor-residing Tregs. Based on this, we developed BAY 3375968, a novel anti-human CCR8 antibody, along with respective surrogate anti-mouse CCR8 antibodies, and demonstrated their in vitro mode-of-action through induction of potent antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) activities. In vivo, anti-mouse CCR8 antibodies effectively depleted Tregs within the TME primarily via ADCP, leading to increased CD8+ T cell infiltration and subsequent tumor growth inhibition across various cancer models. This monotherapeutic efficacy was significantly enhanced in combination with ICIs. Collectively, these findings suggest that CCR8 targeting represents a promising strategy for Treg depletion in cancer therapies. BAY 3375968 is currently under investigation in a Phase I clinical trial (NCT05537740).


Subject(s)
Receptors, CCR8 , T-Lymphocytes, Regulatory , Tumor Microenvironment , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Receptors, CCR8/immunology , Receptors, CCR8/antagonists & inhibitors , Animals , Mice , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , CD8-Positive T-Lymphocytes/immunology , Female , Antibody-Dependent Cell Cytotoxicity , Lymphocyte Depletion , Cell Line, Tumor , Phagocytosis/drug effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use
2.
Cancer Immunol Immunother ; 73(3): 60, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38400933

ABSTRACT

Over the past decade, US Food and Drug Administration (FDA)-approved immune checkpoint inhibitors that target programmed death-1 (PD-1) have demonstrated significant clinical benefit particularly in patients with PD-L1 expressing tumors. Toripalimab is a humanized anti-PD-1 antibody, approved by FDA for first-line treatment of nasopharyngeal carcinoma in combination with chemotherapy. In a post hoc analysis of phase 3 studies, toripalimab in combination with chemotherapy improved overall survival irrespective of PD-L1 status in nasopharyngeal carcinoma (JUPITER-02), advanced non-small cell lung cancer (CHOICE-01) and advanced esophageal squamous cell carcinoma (JUPITER-06). On further characterization, we determined that toripalimab is molecularly and functionally differentiated from pembrolizumab, an anti-PD-1 mAb approved previously for treating a wide spectrum of tumors. Toripalimab, which binds the FG loop of PD-1, has 12-fold higher binding affinity to PD-1 than pembrolizumab and promotes significantly more Th1- and myeloid-derived inflammatory cytokine responses in healthy human PBMCs in vitro. In an ex vivo system employing dissociated tumor cells from treatment naïve non-small cell lung cancer patients, toripalimab induced several unique genes in IFN-γ and immune cell pathways, showed different kinetics of activation and significantly enhanced IFN-γ signature. Additionally, binding of toripalimab to PD-1 induced lower levels of SHP1 and SHP2 recruitment, the negative regulators of T cell activation, in Jurkat T cells ectopically expressing PD-1. Taken together, these data demonstrate that toripalimab is a potent anti-PD-1 antibody with high affinity PD-1 binding, strong functional attributes and demonstrated clinical activity that encourage its continued clinical investigation in several types of cancer.


Subject(s)
Antibodies, Monoclonal, Humanized , Carcinoma, Non-Small-Cell Lung , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lung Neoplasms , Nasopharyngeal Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , B7-H1 Antigen , Programmed Cell Death 1 Receptor , Lung Neoplasms/drug therapy , Nasopharyngeal Carcinoma , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , T-Lymphocytes/pathology
3.
Front Immunol ; 10: 601, 2019.
Article in English | MEDLINE | ID: mdl-31001248

ABSTRACT

Therapeutic antibodies have the potential to induce immunogenicity leading to the development of anti-drug antibodies (ADA) that consequently may result in reduced serum drug concentrations, a loss of efficacy or potential hypersensitivity reactions. Among other factors, aggregated antibodies have been suggested to promote immunogenicity, thus enhancing ADA production. Dendritic cells (DC) are the most efficient antigen-presenting cell population and are crucial for the initiation of T cell responses and the subsequent generation of an adaptive immune response. This work focuses on the development of predictive in vitro assays that can monitor DC maturation, in order to determine whether drug products have direct DC stimulatory capabilities. To this end, four independent laboratories aligned a common protocol to differentiate human monocyte-derived DC (moDC) that were treated with either native or aggregated preparations of infliximab, natalizumab, adalimumab, or rituximab. These drug products were subjected to different forms of physical stress, heat and shear, resulting in aggregation and the formation of subvisible particles. Each partner developed and optimized assays to monitor diverse end-points of moDC maturation: measuring the upregulation of DC activation markers via flow cytometry, analyzing cytokine, and chemokine production via mRNA and protein quantification and identifying cell signaling pathways via quantification of protein phosphorylation. These study results indicated that infliximab, with the highest propensity to form aggregates when heat-stressed, induced a marked activation of moDC as measured by an increase in CD83 and CD86 surface expression, IL-1ß, IL-6, IL-8, IL-12, TNFα, CCL3, and CCL4 transcript upregulation and release of respective proteins, and phosphorylation of the intracellular signaling proteins Syk, ERK1/2, and Akt. In contrast, natalizumab, which does not aggregate under these stress conditions, induced no DC activation in any assay system, whereas adalimumab or rituximab aggregates induced only slight parameter variation. Importantly, the data generated in the different assay systems by each partner site correlated and supported the use of these assays to monitor drug-intrinsic propensities to drive maturation of DC. This moDC assay is also a valuable tool as an in vitro model to assess the intracellular mechanisms that drive DC activation by aggregated therapeutic proteins.


Subject(s)
Antibodies, Monoclonal/pharmacology , Dendritic Cells/drug effects , Biological Assay , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/metabolism , Humans
4.
Methods Mol Biol ; 767: 449-61, 2011.
Article in English | MEDLINE | ID: mdl-21822895

ABSTRACT

Improving our understanding of the interactions between human dendritic cells (DCs) and T cells may contribute to the development of therapeutic strategies for a variety of immune-mediated disorders. The possibility of using DCs themselves as tools to manipulate immune responses opens even greater therapeutic avenues. Current methods of generating human DCs are both inadequate and susceptible to high levels of variability between individuals. DCs differentiated from human embryonic stem cells (hESCs) could provide a more reliable, consistent solution. DCs have now successfully been differentiated from hESCs and more recently this has been repeated using protocols that avoid the inclusion of animal products, an important modification for clinical use. We have developed a novel method for the generation of DCs from hESCs in the absence of animal products that does not necessitate a separate embryoid body (EB) generation step. The technique involves the use of four growth factors and their successive removal from culture, resulting in accumulation of DCs with phenotypic, morphological, and immunostimulatory properties comparable to those of classical human monocyte-derived DCs. In addition to the application of hESC-derived DCs in basic research and novel approaches to cancer immunotherapy, they may also play a central role in the field of regenerative medicine. Tolerogenic DCs differentiated from hESCs may be used to persuade the immune system of the recipients of cell replacement therapy to tolerate allogeneic tissues differentiated from the same hESC line. Such an approach may help to address the immunological barriers that threaten to derail the clinical application of hESCs.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Dendritic Cells/cytology , Embryonic Stem Cells/cytology , Cell Count , Cell Differentiation/drug effects , Cells, Cultured , Collagen/pharmacology , Colony-Forming Units Assay , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Drug Combinations , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Laminin/pharmacology , Monocytes/cytology , Monocytes/drug effects , Proteoglycans/pharmacology
5.
Regen Med ; 6(3): 303-18, 2011 May.
Article in English | MEDLINE | ID: mdl-21548736

ABSTRACT

AIM: Dendritic cell (DC)-based vaccines are designed to exploit the intrinsic capacity of these highly effective antigen presenting cells to prime and boost antigen-specific T-cell immune responses. Successful development of DC-based vaccines will be dependent on the ability to utilize and harness the full potential of these potent immune stimulatory cells. Recent advances to generate DCs derived from human embryonic stem cells (hESCs) that are suitable for clinical use represent an alternative strategy from conventional approaches of using patient-specific DCs. Although the differentiation of hESC-derived DCs in serum-free defined conditions has been established, the stimulatory potential of these hESC-derived DCs have not been fully evaluated. METHODS: hESC-derived DCs were differentiated in serum-free defined culture conditions. The delivery of antigen into hESC-derived DCs was investigated using mRNA transfection and replication-deficient adenoviral vector transduction. hESC-derived DCs modified with antigen were evaluated for their capacity to stimulate antigen-specific T-cell responses with known HLA matching. Since IL-12 is a key cytokine that drives T-cell function, further enhancement of DC potency was evaluated by transfecting mRNA encoding the IL-12p70 protein into hESC-derived DCs. RESULTS: The transfection of mRNA into hESC-derived DCs was effective for heterologous protein expression. The efficiency of adenoviral vector transduction into hESC-derived DCs was poor. These mRNA-transfected DCs were capable of stimulating human telomerase reverse transcriptase antigen-specific T cells composed of varying degrees of HLA matching. In addition, we observed the transfection of mRNA encoding IL-12p70 enhanced the T-cell stimulation potency of hESC-derived DCs. CONCLUSION: These data provide support for the development and modification of hESC-derived DCs with mRNA as a potential strategy for the induction of T-cell-mediated immunity.


Subject(s)
Antigen Presentation/genetics , Antigen Presentation/immunology , Dendritic Cells/immunology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Adenoviridae/drug effects , Adenoviridae/genetics , Antigen Presentation/drug effects , Cell Line , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Embryonic Stem Cells/drug effects , Epitopes/immunology , Gene Expression Regulation/drug effects , Genetic Vectors/genetics , Green Fluorescent Proteins/metabolism , Histocompatibility Testing , Humans , Immunologic Memory/drug effects , Immunologic Memory/immunology , Interferon-gamma/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Peptides/pharmacology , Phenotype , Phosphoproteins/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transduction, Genetic , Transfection , Viral Matrix Proteins/immunology
7.
Regen Med ; 4(4): 513-26, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19580370

ABSTRACT

AIM: Dendritic cell (DC)-based vaccines have a potential utility for use in the treatment of malignancy. Human embryonic stem cells (hESCs) may provide a more cost-effective and reliable source of DCs for immunotherapy purposes, providing on-demand access for patients. METHOD: We developed a protocol to generate DCs from hESCs in vitro in the absence of serum and feeder cells. This protocol uses growth factors bone morphogenetic protein-4, granulocyte macrophage-colony stimulating factor (GM-CSF), stem cell factor and VEGF in serum-free media to generate hESC-derived monocytic cells. These cells are further differentiated to hESC-derived immature DCs with GM-CSF and IL-4, and matured to hESC-derived mature DCs with a maturation cocktail consisting of GM-CSF, TNF-alpha, IL-1beta, IFN-gamma and PGE2. RESULTS: This study demonstrates the applicability of our defined differentiation process in generating functional hESC-derived DCs from multiple hESC lines. We show that hESC-derived immature DCs phagocytose, process, and present antigen upon maturation. hESC-derived mature DCs express the maturation marker CD83, produce Th1-directing cytokine IL-12p70, migrate in response to chemokine, and activate both viral and tumor antigen-specific T-cell responses. CONCLUSION: We developed a chemically defined system to generate unlimited numbers of DCs from hESCs. Our results demonstrate that hESC-derived DCs generated from this process are immunogenic and have the potential to be used for DC immunotherapy.


Subject(s)
Biotechnology/methods , Cell Differentiation/physiology , Dendritic Cells/cytology , Embryonic Stem Cells/cytology , Immunotherapy/methods , Vaccines/biosynthesis , Bone Morphogenetic Protein 4 , Embryonic Stem Cells/physiology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Stem Cell Factor , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Telomerase/metabolism , Vascular Endothelial Growth Factor A
8.
J Biol Chem ; 283(49): 34414-22, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18826951

ABSTRACT

The relationship between intermembrane spacing, adhesion efficiency, and lateral organization of adhesion receptors has not been established for any adhesion system. We have utilized the CD2 ligand CD48 with two (wild type CD48 (CD48-WT)), four (CD48-CD2), or five (CD48-CD22) Ig-like domains. CD48-WT was 10-fold more efficient in mediating adhesion than CD48-CD2 or CD48-CD22. Electron tomography of contact areas with planar bilayers demonstrated average intermembrane spacing of 12.8 nm with CD48-WT, 14.7 nm with CD48-CD2, and 15.6 nm with CD48-CD22. Both CD48-CD2 and CD48-CD22 chimeras segregated completely from CD48-WT in mixed contact areas. In contrast, CD48-CD2 and CD48-CD22 co-localized when mixed contacts were formed. Confocal imaging of immunological synapses formed between primary T lymphocytes and Chinese hamster ovary cells presenting major histocompatibility complex-peptide complexes, and different forms of CD48 demonstrated that CD48-CD2 and CD48-CD22 induce an eccentric CD2/T cell antigen receptor cluster. We propose that this reorganization of the immunological synapse sequesters the T cell antigen receptor in a location where it cannot interact with its ligand and dramatically reduces T cell sensitivity.


Subject(s)
Antigens, CD/chemistry , CD2 Antigens/chemistry , Immunological Synapses/physiology , T-Lymphocytes/metabolism , Animals , CD48 Antigen , CHO Cells , Cell Adhesion , Cell Proliferation , Cricetinae , Cricetulus , Electron Microscope Tomography/methods , Flow Cytometry , Mice , Microscopy, Fluorescence/methods , Sialic Acid Binding Ig-like Lectin 2/chemistry
9.
J Immunol ; 181(7): 4852-63, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802089

ABSTRACT

Short-lived TCR microclusters and a longer-lived protein kinase Ctheta-focusing central supramolecular activation cluster (cSMAC) have been defined in model immunological synapses (IS). In different model systems, CD28-mediated costimulatory interactions have been detected in microclusters, the cSMAC, or segregated from the TCR forming multiple distinct foci. The relationship between TCR and costimulatory molecules in the physiological IS of T cell-dendritic cell (DC) is obscure. To study the dynamic relationship of CD28-CD80 and TCR interactions in the T cell-DC IS during Ag-specific T cell activation, we generated CD80-eCFP mice using bacterial artificial chromosome transgenic technology. In splenic DCs, endogenous CD80 and CD80-eCFP localized to plasma membrane and Golgi apparatus, and CD80-eCFP was functional in vivo. In the OT-II T cell-DC IS, multiple segregated TCR, CD80, and LFA-1 clusters were detected. In the T cell-DC synapse CD80 clusters were colocalized with CD28 and PKCtheta, a characteristic of the cSMAC. Acute blockade of TCR signaling with anti-MHC Ab resulted in a rapid reduction in Ca(2+) signaling and the number and size of the CD80 clusters, a characteristic of TCR microclusters. Thus, the T cell-DC interface contains dynamic costimulatory foci that share characteristics of microclusters and cSMACs.


Subject(s)
B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Dendritic Cells/immunology , Immunological Synapses/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Receptors, Antigen, T-Cell/physiology , T-Lymphocyte Subsets/immunology , Amino Acid Sequence , Animals , B7-1 Antigen/genetics , B7-1 Antigen/physiology , CD28 Antigens/genetics , CD28 Antigens/physiology , CHO Cells , Chromosomes, Artificial, Bacterial/genetics , Cricetinae , Cricetulus , Dendritic Cells/enzymology , Dendritic Cells/metabolism , Green Fluorescent Proteins/genetics , Immunological Synapses/enzymology , Immunological Synapses/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Protein Kinase C-theta , Protein Transport/genetics , Protein Transport/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/metabolism
10.
Curr Opin Immunol ; 18(4): 512-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16777399

ABSTRACT

Dendritic cells (DCs) are myeloid lineage cells that are imprinted by their environment and that mature in response to microbial products. A crucial role of the DC is to impart this context-specific information to T cells as well as to present self and foreign MHC-peptide complexes through formation of an immunological synapse. The structure of the T cell-DC immunological synapse departs from the canonical structure formed with B cells or with supported planar bilayers in that it has multiple foci of T-cell receptor interactions rather than a central focus. Recent studies on model systems provide insight into the mechanisms and biological consequences of the unique T cell-DC synaptic patterns.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , T-Lymphocytes/immunology , Animals , Cell Membrane/immunology , Dendritic Cells/cytology , Humans , Lipid Bilayers/immunology , T-Lymphocytes/cytology
11.
J Immunol ; 175(12): 7829-36, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16339518

ABSTRACT

The binding of costimulatory ligand CD80 to CD28 or CTLA-4 on T cells plays an important role in the regulation of the T cell response. We have examined the role of the cytoplasmic domain of CD80 in murine T cell costimulation and its organization in the immunological synapse (IS). Removal of CD80 cytoplasmic tail decreased its effectiveness in costimulating T cell proliferative response and early IL-2 production in response to agonist MHC-peptide complexes. Immunofluorescent study showed a decreased tailless CD80 accumulation in the IS of naive T cells. The two forms of CD80 accumulated differently at the IS; the tailless CD80 was colocalized with the TCR whereas the full-length CD80 was segregated from the TCR. In addition, we showed that CD80, CD28, and protein kinase Ctheta colocalized in the presence or absence of the CD80 cytoplasmic tail. Thus, the cytoplasmic tail of CD80 regulates its spatial localization at the IS and that of its receptors and T cell signaling molecules such as protein kinase Ctheta, and thereby facilitates full T cell activation.


Subject(s)
B7-1 Antigen/physiology , Cytoplasm/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD , Antigens, Differentiation/metabolism , B7-1 Antigen/chemistry , B7-1 Antigen/metabolism , CD28 Antigens/metabolism , CHO Cells , CTLA-4 Antigen , Cricetinae , Isoenzymes/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Multiprotein Complexes/immunology , Protein Kinase C/metabolism , Protein Kinase C-theta , Protein Structure, Tertiary , Transfection
12.
J Immunol ; 170(4): 1830-8, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574348

ABSTRACT

A human IgM Ab, serum-derived human IgM 12 (sHIgM12), is identified that binds mouse and human dendritic cells (DC), inducing dramatic immunopotentiation following treatment of the mouse DC in vitro. Competition, transfection, and knockout studies identified the ligand on mouse DC as the costimulatory molecule family member B7-DC. Potent T cell responses are stimulated by Ag-pulsed DC treated with the sHIgM12 Ab in vitro and upon adoptive transfer of Ab-treated Ag-pulsed DC into animals. The multivalent structure of pentameric IgM provides the potential for cross-linking cell surface targets, endowing the soluble Abs with biological potential not normally associated with immune function. The ability of the sHIgM12 Ab to potentiate the immune response is dependent on the multimeric structure of IgM, as bivalent monomers do not retain this property. Furthermore, pretreatment of DC with IgM monomers blocks subsequent potentiation by intact IgM pentamers, an indication that cross-linking of B7-DC on the cell surface is critical for potentiation of Ag presentation. These findings imply that, in addition to known costimulatory roles, B7-DC can function as a receptor for signals delivered by cells expressing B7-DC ligands.


Subject(s)
Adjuvants, Immunologic/metabolism , B7-1 Antigen/metabolism , Binding Sites, Antibody , Dendritic Cells/immunology , Immunoglobulin M/blood , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/physiology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/physiology , Antigen Presentation/immunology , Binding Sites, Antibody/physiology , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Humans , Immunity, Innate , Immunoglobulin M/chemistry , Immunoglobulin M/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Programmed Cell Death 1 Ligand 2 Protein , Receptors, Fc/metabolism , Receptors, IgG/metabolism , Species Specificity , Structure-Activity Relationship
13.
Curr Opin Cell Biol ; 14(5): 575-80, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12231352

ABSTRACT

Naïve T cell activation requires the interactions of antigen receptors, adhesion molecules and co-stimulatory molecules. Antigen receptors and adhesion molecules are involved in spatio-temporal movement to form a stable immunological synapse. This stable junction interrupts T cell migration, and provides a platform for temporally regulated co-stimulatory receptor signaling spanning a period of days.


Subject(s)
Lymphocyte Activation , Signal Transduction , T-Lymphocytes/physiology , Animals , Cell Adhesion , Cell Differentiation , Cell Division , Cell Movement , Humans , Lymph Nodes/metabolism , Synapses , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...