Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
JCI Insight ; 3(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29415897

ABSTRACT

NK cell-based immunotherapies have been gaining traction in the clinic for treatment of cancer. IL-15 is currently being used in number of clinical trials to improve NK cell expansion and function. The objective of this study is to evaluate the effect of repetitive IL-15 exposure on NK cells. An in vitro model in which human NK cells are continuously (on on on) or intermittently (on off on) treated with IL-15 was used to explore this question. After treatment, cells were evaluated for proliferation, survival, cell cycle gene expression, function, and metabolic processes. Our data indicate that continuous treatment of NK cells with IL-15 resulted in decreased viability and a cell cycle arrest gene expression pattern. This was associated with diminished signaling, decreased function both in vitro and in vivo, and reduced tumor control. NK cells continuously treated with IL-15 also displayed a reduced mitochondrial respiration profile when compared with NK cells treated intermittently with IL-15. This profile was characterized by a decrease in the spare respiratory capacity that was dependent on fatty acid oxidation (FAO). Limiting the strength of IL-15 signaling via utilization of an mTOR inhibitor rescued NK cell functionality in the group continuously treated with IL-15. The findings presented here show that human NK cells continuously treated with IL-15 undergo a process consistent with exhaustion that is accompanied by a reduction in FAO. These findings should inform IL-15-dosing strategies in NK cell cancer immunotherapeutic settings.


Subject(s)
Fatty Acids/metabolism , Immunotherapy/methods , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Neoplasms/therapy , Animals , Blood Buffy Coat/cytology , Cell Line, Tumor , Clinical Trials as Topic , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Mice , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction/drug effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Whole-Body Irradiation , Xenograft Model Antitumor Assays
3.
Cancer Res Treat ; 49(4): 1140-1152, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28231426

ABSTRACT

PURPOSE: The selective elimination of cancer stem cells (CSCs) in tumor patients is a crucial goal because CSCs cause drug refractory relapse. To improve the current conventional bispecific immune-engager platform, a 16133 bispecific natural killer (NK) cell engager (BiKE), consisting of scFvs binding FcγRIII (CD16) on NK cells and CD133 on carcinoma cells, was first synthesized and a modified interleukin (IL)-15 crosslinker capable of stimulating NK effector cells was introduced. MATERIALS AND METHODS: DNA shuffling and ligation techniques were used to assemble and synthesize the 1615133 trispecific NK cell engager (TriKE). The construct was tested for its specificity using flow cytometry, cytotoxic determinations using chromium release assays, and lytic degranulation. IL-15-mediated expansion was measured using flow-based proliferation assays. The level of interferon (IFN)-γ release was measured because of its importance in the anti-cancer response. RESULTS: 1615133 TriKE induced NK cell-mediated cytotoxicity and NK expansion far greater than that achieved with BiKE devoid of IL-15. The drug binding and induction of cytotoxic degranulation was CD133+ specific and the anti-cancer activity was improved by integrating the IL-15 cross linker. The NK cell-related cytokine release measured by IFN-γ detection was higher than that of BiKE. NK cytokine release studies showed that although the IFN-γ levels were elevated, they did not approach the levels achieved with IL-12/IL-18, indicating that release was not at the supraphysiologic level. CONCLUSION: 1615133 TriKE enhances the NK cell anti-cancer activity and provides a self-sustaining mechanism via IL-15 signaling. By improving the NK cell performance, the new TriKE represents a highly active drug against drug refractory relapse mediated by CSCs.


Subject(s)
AC133 Antigen/antagonists & inhibitors , Antibody-Dependent Cell Cytotoxicity/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology , T-Cell Antigen Receptor Specificity/immunology , Cell Culture Techniques , Cell Degranulation/immunology , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Genetic Engineering , Humans , Immunotherapy , Interferon-gamma/biosynthesis , Interleukin-15/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism
4.
Clin Cancer Res ; 22(14): 3440-50, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26847056

ABSTRACT

PURPOSE: The effectiveness of NK cell infusions to induce leukemic remission is limited by lack of both antigen specificity and in vivo expansion. To address the first issue, we previously generated a bispecific killer engager (BiKE) containing single-chain scFv against CD16 and CD33 to create an immunologic synapse between NK cells and CD33(+) myeloid targets. We have now incorporated a novel modified human IL15 crosslinker, producing a 161533 trispecific killer engager (TriKE) to induce expansion, priming, and survival, which we hypothesize will enhance clinical efficacy. EXPERIMENTAL DESIGN: Reagents were tested in proliferation and functional assays and in an in vivo xenograft model of AML. RESULTS: When compared with the 1633 BiKE, the 161533 TriKE induced superior NK cell cytotoxicity, degranulation, and cytokine production against CD33(+) HL-60 targets and increased NK survival and proliferation. Specificity was shown by the ability of a 1615EpCAM TriKE to kill CD33-EpCAM(+) targets. Using NK cells from patients after allogeneic stem cell transplantation when NK cell function is defective, the 161533 TriKE restored potent NK function against primary AML targets and induced specific NK cell proliferation. These results were confirmed in an immunodeficient mouse HL-60-Luc tumor model where the 161533 TriKE exhibited superior antitumor activity and induced in vivo persistence and survival of human NK cells for at least 3 weeks. CONCLUSIONS: Off-the-shelf 161533 TriKE imparts antigen specificity and promotes in vivo persistence, activation, and survival of NK cells. These qualities are ideal for NK cell therapy of myeloid malignancies or targeting antigens of solid tumors. Clin Cancer Res; 22(14); 3440-50. ©2016 AACRSee related commentary by Talmadge, p. 3419.


Subject(s)
Interleukin-15/immunology , Killer Cells, Natural/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Animals , Cell Proliferation/physiology , Cytotoxicity, Immunologic/immunology , Epithelial Cell Adhesion Molecule/immunology , HL-60 Cells , HT29 Cells , Humans , Lymphocyte Activation/immunology , Mice , Stem Cells/immunology , Transplantation, Homologous/methods
5.
Blood ; 123(19): 3016-26, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24652987

ABSTRACT

Myelodysplastic syndromes (MDS) are stem cell disorders that can progress to acute myeloid leukemia. Although hematopoietic cell transplantation can be curative, additional therapies are needed for a disease that disproportionally afflicts the elderly. We tested the ability of a CD16xCD33 BiKE to induce natural killer (NK) cell function in 67 MDS patients. Compared with age-matched normal controls, CD7(+) lymphocytes, NK cells, and CD16 expression were markedly decreased in MDS patients. Despite this, reverse antibody-dependent cell-mediated cytotoxicity assays showed potent degranulation and cytokine production when resting MDS-NK cells were triggered with an agonistic CD16 monoclonal antibody. Blood and marrow MDS-NK cells treated with bispecific killer cell engager (BiKE) significantly enhanced degranulation and tumor necrosis factor-α and interferon-γ production against HL-60 and endogenous CD33(+) MDS targets. MDS patients had a significantly increased proportion of immunosuppressive CD33(+) myeloid-derived suppressor cells (MDSCs) that negatively correlated with MDS lymphocyte populations and CD16 loss on NK cells. Treatment with the CD16xCD33 BiKE successfully reversed MDSC immunosuppression of NK cells and induced MDSC target cell lysis. Lastly, the BiKE induced optimal MDS-NK cell function irrespective of disease stage. Our data suggest that the CD16xCD33 BiKE functions against both CD33(+) MDS and MDSC targets and may be therapeutically beneficial for MDS patients.


Subject(s)
Antibodies, Bispecific/immunology , Killer Cells, Natural/immunology , Myelodysplastic Syndromes/immunology , Receptors, IgG/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Bispecific/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Cells, Cultured , Female , Flow Cytometry , HL-60 Cells , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Male , Middle Aged , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/prevention & control , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL