Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 10(9)2022 09.
Article in English | MEDLINE | ID: mdl-36162918

ABSTRACT

BACKGROUND: The tumor microenvironment contains stromal cells, including endothelial cells and fibroblasts, that aid tumor growth and impair immune cell function. Many solid tumors remain difficult to cure because of tumor-promoting stromal cells, but current therapies targeting tumor stromal cells are constrained by modest efficacy and toxicities. TEM8 is a surface antigen selectively upregulated on tumor and tumor stromal cells, endothelial cells and fibroblasts that may be targeted with specific natural killer (NK) cell engagement. METHODS: A Tri-specific Killer Engager (TriKE) against TEM8-'cam1615TEM8'-was generated using a mammalian expression system. Its function on NK cells was assessed by evaluation of degranulation, inflammatory cytokine production, and killing against tumor and stroma cell lines in standard co-culture and spheroid assays. cam1615TEM8-mediated proliferation and STAT5 phosphorylation in NK cells was tested and compared with T cells by flow cytometry. NK cell proliferation, tumor infiltration, and tumor and tumor-endothelium killing by cam1615TEM8 and interleukin-15 (IL-15) were assessed in NOD scid gamma (NSG) mice. RESULTS: cam1615TEM8 selectively stimulates NK cell degranulation and inflammatory cytokine production against TEM8-expressing tumor and stromal cell lines. The increased activation translated to superior NK cell killing of TEM8-expressing tumor spheroids. cam1615TEM8 selectively stimulated NK cell but not T cell proliferation in vitro and enhanced NK cell proliferation, survival, and tumor infiltration in vivo. Finally, cam1615TEM8 stimulated NK cell killing of tumor and tumor endothelial cells in vivo. CONCLUSIONS: Our findings indicate that the cam1615TEM8 TriKE is a novel anti-tumor, anti-stroma, and anti-angiogenic cancer therapy for patients with solid tumors. This multifunctional molecule works by selectively targeting and activating NK cells by costimulation with IL-15, and then targeting that activity to TEM8+ tumor cells and TEM8+ tumor stroma.


Subject(s)
Interleukin-15 , Neoplasms , Animals , Antigens, Surface/metabolism , Endothelial Cells , Interleukin-15/metabolism , Killer Cells, Natural , Mammals/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Microfilament Proteins , Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, Cell Surface , STAT5 Transcription Factor/metabolism , Tumor Microenvironment
2.
Cancer Immunol Res ; 8(9): 1139-1149, 2020 09.
Article in English | MEDLINE | ID: mdl-32661096

ABSTRACT

Natural killer (NK) cells are potent immune modulators that can quickly lyse tumor cells and elicit inflammatory responses. These characteristics make them ideal candidates for immunotherapy. However, unlike T cells, NK cells do not possess clonotypic receptors capable of specific antigen recognition and cannot expand via activating receptor signals alone. To enable NK cells with these capabilities, we created and have previously described a tri-specific killer engager (TriKE) platform capable of inducing antigen specificity and cytokine-mediated NK-cell expansion. TriKE molecules have three arms: (i) a single-chain variable fragment (scFv) against the activating receptor CD16 on NK cells to trigger NK-cell activation, (ii) an scFv against a tumor-associated antigen (CD33 here) to induce specific tumor target recognition, and (iii) an IL15 moiety to trigger NK-cell expansion and priming. Here, we demonstrate that by modifying the anti-CD16 scFv with a humanized single-domain antibody against CD16, we improved TriKE functionality. A CD33-targeting second-generation TriKE induced stronger and more specific NK-cell proliferation without T-cell stimulation, enhanced in vitro NK-cell activation and killing of CD33-expressing targets, and improved tumor control in preclinical mouse models. Given these improved functional characteristics, we propose rapid translation of second-generation TriKEs into the clinic.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-15/administration & dosage , Interleukin-15/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Animals , Disease Models, Animal , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
3.
Methods Mol Biol ; 1441: 333-46, 2016.
Article in English | MEDLINE | ID: mdl-27177679

ABSTRACT

Cancer immunotherapies have gained significant momentum over the past decade, particularly with the advent of checkpoint inhibitors and CAR T-cells. While the latter personalized targeted immunotherapy has revolutionized the field, a need for off-the-shelf therapies remains. The ability of NK cells to quickly lyse antibody-coated tumors and potently secrete cytokines without prior priming has made NK cells ideal candidates for antigen-specific immunotherapy. NK cells have been targeted to tumors through two main strategies: mono-specific antibodies and bi/tri-specific antibodies. Mono-specific antibodies drive NK cell antibody-dependent cell-mediated cytotoxicity (ADCC) of tumor cells. Bi/tri-specific antibodies drive re-directed lysis of tumor cells through binding of a tumor antigen and direct binding and crosslinking of the CD16 receptor on NK cells, thus bypassing the need for binding of the Fc portion of mono-specific antibodies. This chapter focuses on the generation of bi- and tri-specific killer engagers (BiKEs and TriKEs) meant to target NK cells to tumors. BiKEs and TriKEs are smaller molecules composed of 2-3 variable portions of antibodies with different specificities, and represent a novel and more versatile strategy compared to traditional bi- and tri-specific antibody platforms.


Subject(s)
Antibodies, Bispecific/metabolism , Antibodies, Monoclonal/metabolism , Killer Cells, Natural/immunology , Cell Line, Tumor , Cells, Cultured , Humans , Immunotherapy, Adoptive , Killer Cells, Natural/transplantation , Neoplasms/immunology , Neoplasms/therapy
4.
J Immunol ; 193(7): 3344-54, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25172483

ABSTRACT

The Notch signaling pathway plays a substantial role in human NK cell development. However, the role of Notch on killer Ig-like receptor (KIR) upregulation and acquisition of effector function has not been explored. To evaluate how Notch influences terminal differentiation, cord blood-derived NK cells or sorted peripheral blood NK cells were cultured with IL-15 for 7 d with inhibitory or activating Notch signals. Inhibition of Notch signaling significantly decreased KIR expression, whereas activation enhanced it. Overexpression of activated Notch on cord blood-derived NK cells resulted in a 2-fold increase in KIR expression, indicating that Notch signaling plays a direct, cell-intrinsic role in KIR regulation. Moreover, Notch-mediated KIR expression on NK cells is regulated through cis inhibition by delta-like ligand 1. Notch signaling also enhances CD16 upregulation that precedes KIR expression. Concomitant with the upregulation of KIR and CD16, Notch signaling induces increased cytolytic effector capacity and cytokine secretion, even in posttransplant samples in which NK cell function is inherently defective. Given these attributes of Notch signaling, we propose that Notch agonists may enhance NK cell maturation and tumor killing in a posttransplant setting.


Subject(s)
Gene Expression Regulation/immunology , Killer Cells, Natural/immunology , Receptors, KIR/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , Calcium-Binding Proteins , Cytokines/immunology , Female , GPI-Linked Proteins/immunology , Humans , Intercellular Signaling Peptides and Proteins/immunology , K562 Cells , Killer Cells, Natural/cytology , Male , Membrane Proteins/immunology , Receptors, IgG/immunology
5.
J Exp Med ; 211(6): 1079-91, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24842371

ABSTRACT

Autosomal recessive mutations in UNC13D, the gene that encodes Munc13-4, are associated with familial hemophagocytic lymphohistiocytosis type 3 (FHL3). Munc13-4 expression is obligatory for exocytosis of lytic granules, facilitating cytotoxicity by T cells and natural killer (NK) cells. The mechanisms regulating Munc13-4 expression are unknown. Here, we report that Munc13-4 is highly expressed in differentiated human NK cells and effector CD8(+) T lymphocytes. A UNC13D c.118-308C>T mutation, causative of FHL3, disrupted binding of the ETS family member ELF1 to a conserved intronic sequence. This mutation impairs UNC13D intron 1 recruitment of STAT4 and the chromatin remodeling complex component BRG1, diminishing active histone modifications at the locus. The intronic sequence acted as an overall enhancer of Munc13-4 expression in cytotoxic lymphocytes in addition to representing an alternative promoter encoding a novel Munc13-4 isoform. Mechanistically, T cell receptor engagement facilitated STAT4-dependent Munc13-4 expression in naive CD8(+) T lymphocytes. Collectively, our data demonstrates how chromatin remodeling within an evolutionarily conserved regulatory element in intron 1 of UNC13D regulates the induction of Munc13-4 expression in cytotoxic lymphocytes and suggests that an alternative Munc13-4 isoform is required for lymphocyte cytotoxicity. Thus, mutations associated with primary immunodeficiencies may cause disease by disrupting transcription factor binding.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Deficiency Syndromes/immunology , Killer Cells, Natural/immunology , Membrane Proteins/immunology , Point Mutation/immunology , Blotting, Western , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , DNA Helicases/genetics , DNA Helicases/immunology , DNA Helicases/metabolism , Gene Expression Regulation/immunology , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Introns/genetics , Killer Cells, Natural/metabolism , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Protein Binding/genetics , Protein Binding/immunology , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , STAT4 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
6.
J Immunol ; 192(8): 3889-97, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24634493

ABSTRACT

Acquisition of a functional NK cell repertoire, known as education or licensing, is a complex process mediated through inhibitory receptors that recognize self. We found that NK cells containing self-killer Ig-like receptors for cognate HLA ligand in vivo were less susceptible to apoptosis. In vitro IL-15 withdrawal showed that uneducated NK cells upregulated Bim and Fas. Conversely, educated NK cells upregulated Fas ligand (FasL) under these conditions. Induction of cell death and Bim expression on uneducated cells correlated with increased IL-2Rα expression. Overexpression and knockdown studies showed that higher IL-2Rα limits NK cell survival in a novel manner that is independent from the role of IL-2 in activation-induced cell death. To study the role of FasL in induction of IL-2Rα(hi) NK cell death, a coculture assay with FasL-blocking Abs was used. IL-15 withdrawal led to FasL-dependent killing of IL-2Rα(hi) NK cells by more educated IL-2Rα(lo) NK cells. Finally, CMV reactivation induces a potent long-lasting population of licensed NK cells with enhanced survival. These findings show that education-dependent NK cell survival advantages and killing of uneducated NK cells result in the maintenance of a functional repertoire, which may be manipulated to exploit NK cells for cancer immunotherapy.


Subject(s)
Fas Ligand Protein/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Killer Cells, Natural/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Communication , Cell Differentiation , Cell Proliferation , Cell Survival/drug effects , Cell Survival/immunology , Cytomegalovirus/physiology , Fas Ligand Protein/genetics , Gene Expression Regulation/drug effects , Homeostasis , Humans , Interleukin-15/pharmacology , Interleukin-2 Receptor alpha Subunit/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, KIR/genetics , Receptors, KIR/metabolism , Virus Activation
7.
Blood ; 119(13): 3064-72, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22323453

ABSTRACT

NK-cell function is regulated by the integration of signals received from activating and inhibitory receptors. Here we show that a novel immune receptor, T-cell Ig and mucin-containing domain-3 (Tim-3), is expressed on resting human NK cells and is up-regulated on activation. The NK92 NK-cell line engineered to overexpress Tim-3 showed a marked increase in IFN-γ production in the presence of soluble rhGal-9 or Raji tumor cells engineered to express Gal-9. The Tim-3(+) population of low-dose IL-12/IL-18-activated primary NK cells significantly increased IFN-γ production in response to soluble rhGal-9, Gal-9 presented by cell lines, and primary acute myelogenous leukemia (AML) targets that endogenously express Gal-9. This effect is highly specific as Tim-3 Ab blockade significantly decreased IFN-γ production, and Tim-3 cross-linking induced ERK activation and degradation of IκBα. Exposure to Gal-9-expressing target cells had little effect on CD107a degranulation. Reconstituted NK cells obtained from patients after hematopoietic cell transplantation had diminished expression of Tim-3 compared with paired donors. This observation correlates with the known IFN-γ defect seen early posttransplantation. In conclusion, we show that Tim-3 functions as a human NK-cell coreceptor to enhance IFN-γ production, which has important implications for control of infectious disease and cancer.


Subject(s)
Galectins/pharmacology , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Membrane Proteins/genetics , Adult , Cells, Cultured , Galectins/genetics , Galectins/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HEK293 Cells , Hematopoietic Stem Cell Transplantation , Hepatitis A Virus Cellular Receptor 2 , Humans , Interferon-gamma/blood , Jurkat Cells , Leukemia/blood , Leukemia/genetics , Leukemia/immunology , Leukemia/therapy , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Membrane Proteins/physiology , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism , Receptors, Natural Killer Cell/physiology , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...