1
|
Weiss L, Schluck M, Classens R, de Jonge PKJD, van der Waart A, Nguyen KG, Nguyen TT, Zaharoff DA, Malmberg KJ, Dolstra H, Figdor CG, Sohlberg E, Hammink R. Interleukin-12 decorated nanosized semiflexible Immunofilaments enable directed targeting and augmented IFNγ responses of natural killer cells. Acta Biomater 2025; 191:386-397. [PMID: 39528061 DOI: 10.1016/j.actbio.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Immunotherapies are a powerful strategy to treat cancer by modulating the immune system to raise an anti-tumor immune response. A prime example of immunotherapies are cytokines - small immunomodulatory molecules that are widely used to stimulate immune cells. Undirected administration of cytokines, however, can cause severe side effects, preventing the use of potent cytokines, such as Interleukin (IL)-12, which induces IFNγ responses by cytotoxic effector lymphocytes, including NK cells. Biomaterials, like nanoparticles, can encapsulate IL-12 and accumulate at the tumor site to alleviate side effects. Yet, the released IL-12 might not be directly targeted to extracellular IL-12 receptors on the specific effector cells, thereby potentially compromising the cytokine's therapeutic efficacy. Here, we develop a polymer-based platform to target NK cells, which we call immunofilaments. Immunofilaments are nanosized linear polymers that present an anti-CD16 antibody and IL-12 effectively to NK cells and lead to synergistic NK cell activation as highlighted by an increase in TNFα and IFNγ production and upregulation of multiple activation markers, including CD25, CD69, and degranulation marker CD107a. NK cell proliferation is enhanced in the presence of both anti-CD16 antibody and IL-12 compared to giving IL-12 separately. Finally, we demonstrate that the IF platform is suitable for in vivo applications, as immunofilaments readily activate human NK cells upon administration to mice. STATEMENT OF SIGNIFICANCE: IL-12 is a potent cytokine that stimulates IFNγ responses in NK cells, which supports an anti-tumor immune response. Due to its high potency, the delivery of IL-12 needs to be highly controlled to prevent severe adverse side effects, which can be achieved by using biomaterials. This study shows that nanosized polymers termed Immunofilaments can be used to immobilize IL-12 and effectively target and activate NK cells by co-conjugation of anti-CD16 antibodies. This work is a prime example of careful engineering of innovative biomaterials to improve immunotherapy.
Collapse
Affiliation(s)
- Lea Weiss
- Department of Medical BioSciences, Radboudumc, Geert Grooteplein 26, Nijmegen, GA 6525, the Netherlands; Institute for Chemical Immunology, Nijmegen, GA 6525, the Netherlands; Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, GA 6525, Netherlands
| | - Marjolein Schluck
- Department of Medical BioSciences, Radboudumc, Geert Grooteplein 26, Nijmegen, GA 6525, the Netherlands; Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, GA 6525, Netherlands
| | - René Classens
- Department of Medical BioSciences, Radboudumc, Geert Grooteplein 26, Nijmegen, GA 6525, the Netherlands
| | - Paul K J D de Jonge
- Laboratory of Hematology, Department of Laboratory Medicine, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, GA 6525, USA
| | - Anniek van der Waart
- Laboratory of Hematology, Department of Laboratory Medicine, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, GA 6525, USA
| | - Khue G Nguyen
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill & North Carolina State University, Raleigh, NC, USA
| | - Tam T Nguyen
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill & North Carolina State University, Raleigh, NC, USA
| | - David A Zaharoff
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill & North Carolina State University, Raleigh, NC, USA
| | - Karl-Johan Malmberg
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; The Precision Immunotherapy Alliance, The University of Oslo, Oslo, Norway
| | - Harry Dolstra
- Laboratory of Hematology, Department of Laboratory Medicine, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, GA 6525, USA
| | - Carl G Figdor
- Department of Medical BioSciences, Radboudumc, Geert Grooteplein 26, Nijmegen, GA 6525, the Netherlands; Institute for Chemical Immunology, Nijmegen, GA 6525, the Netherlands; Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, GA 6525, Netherlands.
| | - Ebba Sohlberg
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Roel Hammink
- Department of Medical BioSciences, Radboudumc, Geert Grooteplein 26, Nijmegen, GA 6525, the Netherlands; Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, GA 6525, Netherlands.
| |
Collapse
|
2
|
Bryan AM, You JK, Li G, Kim J, Singh A, Morstein J, Trauner D, Pereira de Sá N, Normile TG, Farnoud AM, London E, Del Poeta M. Cholesterol and sphingomyelin are critical for Fcγ receptor-mediated phagocytosis of Cryptococcus neoformans by macrophages. J Biol Chem 2021; 297:101411. [PMID: 34793834 PMCID: PMC8661020 DOI: 10.1016/j.jbc.2021.101411] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 12/21/2022] Open
Abstract
Cryptococcus neoformans is a fungal pathogen that causes life-threatening meningoencephalitis in lymphopenic patients. Pulmonary macrophages comprise the first line of host defense upon inhalation of fungal spores by aiding in clearance but can also potentially serve as a niche for their dissemination. Given that macrophages play a key role in the outcome of a cryptococcal infection, it is crucial to understand factors that mediate phagocytosis of C. neoformans. Since lipid rafts (high-order plasma membrane domains enriched in cholesterol and sphingomyelin [SM]) have been implicated in facilitating phagocytosis, we evaluated whether these ordered domains govern macrophages' ability to phagocytose C. neoformans. We found that cholesterol or SM depletion resulted in significantly deficient immunoglobulin G (IgG)-mediated phagocytosis of fungus. Moreover, repletion of macrophage cells with a raft-promoting sterol (7-dehydrocholesterol) rescued this phagocytic deficiency, whereas a raft-inhibiting sterol (coprostanol) significantly decreased IgG-mediated phagocytosis of C. neoformans. Using a photoswitchable SM (AzoSM), we observed that the raft-promoting conformation (trans-AzoSM) resulted in efficient phagocytosis, whereas the raft-inhibiting conformation (cis-AzoSM) significantly but reversibly blunted phagocytosis. We observed that the effect on phagocytosis may be facilitated by Fcγ receptor (FcγR) function, whereby IgG immune complexes crosslink to FcγRIII, resulting in tyrosine phosphorylation of FcR γ-subunit (FcRγ), an important accessory protein in the FcγR signaling cascade. Correspondingly, cholesterol or SM depletion resulted in decreased FcRγ phosphorylation. Repletion with 7-dehydrocholesterol restored phosphorylation, whereas repletion with coprostanol showed FcRγ phosphorylation comparable to unstimulated cells. Together, these data suggest that lipid rafts are critical for facilitating FcγRIII-mediated phagocytosis of C. neoformans.
Collapse
Affiliation(s)
- Arielle M Bryan
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Jeehyun Karen You
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Guangtao Li
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - JiHyun Kim
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Ashutosh Singh
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Johannes Morstein
- Department of Chemistry, New York University, New York, New York, USA
| | - Dirk Trauner
- Department of Chemistry, New York University, New York, New York, USA
| | - Nívea Pereira de Sá
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Tyler G Normile
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Amir M Farnoud
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Erwin London
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA; Division of Infectious Diseases, Stony Brook University, Stony Brook, New York, USA; Veteran Affairs Medical Center, Northport, New York, USA.
| |
Collapse
|
3
|
Li W, Li M, Anthony SM, Yu Y. Spatial organization of FcγR and TLR2/1 on phagosome membranes differentially regulates their synergistic and inhibitory receptor crosstalk. Sci Rep 2021; 11:13430. [PMID: 34183758 PMCID: PMC8238967 DOI: 10.1038/s41598-021-92910-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/14/2021] [Indexed: 11/09/2022] Open
Abstract
Many innate immune receptors function collaboratively to detect and elicit immune responses to pathogens, but the physical mechanisms that govern the interaction and signaling crosstalk between the receptors are unclear. In this study, we report that the signaling crosstalk between Fc gamma receptor (FcγR) and Toll-like receptor (TLR)2/1 can be overall synergistic or inhibitory depending on the spatial proximity between the receptor pair on phagosome membranes. Using a geometric manipulation strategy, we physically altered the spatial distribution of FcγR and TLR2 on single phagosomes. We demonstrate that the signaling synergy between FcγR and TLR2/1 depends on the proximity of the receptors and decreases as spatial separation between them increases. However, the inhibitory effect from FcγRIIb on TLR2-dependent signaling is always present and independent of receptor proximity. The overall cell responses are an integration from these two mechanisms. This study presents quantitative evidence that the nanoscale proximity between FcγR and TLR2 functions as a key regulatory mechanism in their signaling crosstalk.
Collapse
Affiliation(s)
- Wenqian Li
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA.,Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, 47405, USA
| | - Miao Li
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA
| | - Stephen M Anthony
- Department of Computational Biology and Biophysics, Sandia National Laboratories, Albuquerque, NM, 87123, USA
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
4
|
Suarez-Kelly L, Sun SH, Ren C, Rampersaud IV, Albertson D, Duggan MC, Noel TC, Courtney N, Buteyn NJ, Moritz C, Yu L, Yildiz VO, Butchar JP, Tridandapani S, Rampersaud AA, Carson WE. Antibody Conjugation of Fluorescent Nanodiamonds for Targeted Innate Immune Cell Activation. ACS APPLIED NANO MATERIALS 2021; 4:3122-3139. [PMID: 34027313 PMCID: PMC8136585 DOI: 10.1021/acsanm.1c00256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
BACKGROUND fluorescent nanodiamonds (FND) are nontoxic, infinitely photostable nanoparticles that emit near-infrared fluorescence and have a modifiable surface allowing for the generation of protein-FND conjugates. FND-mediated immune cell targeting may serve as a strategy to visualize immune cells and promote immune cell activation. METHODS uncoated-FND (uFND) were fabricated, coated with glycidol (gFND), and conjugated with immunoglobulin G (IgG-gFND). In vitro studies were performed using a breast cancer/natural killer/monocyte co-culture system, and in vivo studies were performed using a breast cancer mouse model. RESULTS in vitro studies demonstrated the targeted immune cell uptake of IgG-gFND, resulting in significant immune cell activation and no compromise in immune cell viability. IgG-gFND remained at the tumor site following intratumoral injection compared to uFND which migrated to the liver and kidneys. CONCLUSION antibody-conjugated FND may serve as immune drug delivery vehicles with "track and trace capabilities" to promote directed antitumor activity and minimize systemic toxicities.
Collapse
Affiliation(s)
- Lorena
P. Suarez-Kelly
- The
Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
| | - Steven H. Sun
- Department
of Surgery, The Ohio State University, Columbus, Ohio 43210, United States
| | - Casey Ren
- The
Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
| | - Isaac V. Rampersaud
- Columbus
NanoWorks, Inc., 1507
Chambers Road, Columbus, Ohio 43212, United
States
| | - David Albertson
- Columbus
NanoWorks, Inc., 1507
Chambers Road, Columbus, Ohio 43212, United
States
| | - Megan C. Duggan
- The
Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
| | - Tiffany C. Noel
- The
Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nicholas Courtney
- The
Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathaniel J. Buteyn
- Division
of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Charles Moritz
- Columbus
NanoWorks, Inc., 1507
Chambers Road, Columbus, Ohio 43212, United
States
| | - Lianbo Yu
- Department
of Biomedical Informatics, The Ohio State
University, Columbus, Ohio 43210, United States
| | - Vedat O. Yildiz
- Department
of Biomedical Informatics, The Ohio State
University, Columbus, Ohio 43210, United States
| | - Jonathan P. Butchar
- Division
of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Susheela Tridandapani
- Division
of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Arfaan A. Rampersaud
- Columbus
NanoWorks, Inc., 1507
Chambers Road, Columbus, Ohio 43212, United
States
| | - William E. Carson
- The
Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
- Department
of Surgery, The Ohio State University, Columbus, Ohio 43210, United States
- . Phone: (614)
293-6306. Fax: (614) 293-3465
| |
Collapse
|
5
|
Zhang T, Hu W, Chen W. Plasma Membrane Integrates Biophysical and Biochemical Regulation to Trigger Immune Receptor Functions. Front Immunol 2021; 12:613185. [PMID: 33679752 PMCID: PMC7933204 DOI: 10.3389/fimmu.2021.613185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/06/2021] [Indexed: 11/23/2022] Open
Abstract
Plasma membrane provides a biophysical and biochemical platform for immune cells to trigger signaling cascades and immune responses against attacks from foreign pathogens or tumor cells. Mounting evidence suggests that the biophysical-chemical properties of this platform, including complex compositions of lipids and cholesterols, membrane tension, and electrical potential, could cooperatively regulate the immune receptor functions. However, the molecular mechanism is still unclear because of the tremendous compositional complexity and spatio-temporal dynamics of the plasma membrane. Here, we review the recent significant progress of dynamical regulation of plasma membrane on immune receptors, including T cell receptor, B cell receptor, Fc receptor, and other important immune receptors, to proceed mechano-chemical sensing and transmembrane signal transduction. We also discuss how biophysical-chemical cues couple together to dynamically tune the receptor's structural conformation or orientation, distribution, and organization, thereby possibly impacting their in-situ ligand binding and related signal transduction. Moreover, we propose that electrical potential could potentially induce the biophysical-chemical coupling change, such as lipid distribution and membrane tension, to inevitably regulate immune receptor activation.
Collapse
Affiliation(s)
- Tongtong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Hu
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, State Key Laboratory for Modern Optical Instrumentation, College of Biomedical Engineering and Instrument Science, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Kara S, Amon L, Lühr JJ, Nimmerjahn F, Dudziak D, Lux A. Impact of Plasma Membrane Domains on IgG Fc Receptor Function. Front Immunol 2020; 11:1320. [PMID: 32714325 PMCID: PMC7344230 DOI: 10.3389/fimmu.2020.01320] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
Lipid cell membranes not only represent the physical boundaries of cells. They also actively participate in many cellular processes. This contribution is facilitated by highly complex mixtures of different lipids and incorporation of various membrane proteins. One group of membrane-associated receptors are Fc receptors (FcRs). These cell-surface receptors are crucial for the activity of most immune cells as they bind immunoglobulins such as immunoglobulin G (IgG). Based on distinct mechanisms of IgG binding, two classes of Fc receptors are now recognized: the canonical type I FcγRs and select C-type lectin receptors newly referred to as type II FcRs. Upon IgG immune complex induced cross-linking, these receptors are known to induce a multitude of cellular effector responses in a cell-type dependent manner, including internalization, antigen processing, and presentation as well as production of cytokines. The response is also determined by specific intracellular signaling domains, allowing FcRs to either positively or negatively modulate immune cell activity. Expression of cell-type specific combinations and numbers of receptors therefore ultimately sets a threshold for induction of effector responses. Mechanistically, receptor cross-linking and localization to lipid rafts, i.e., organized membrane microdomains enriched in intracellular signaling proteins, were proposed as major determinants of initial FcR activation. Given that immune cell membranes might also vary in their lipid compositions, it is reasonable to speculate, that the cell membrane and especially lipid rafts serve as an additional regulator of FcR activity. In this article, we aim to summarize the current knowledge on the interplay of lipid rafts and IgG binding FcRs with a focus on the plasma membrane composition and receptor localization in immune cells, the proposed mechanisms underlying this localization and consequences for FcR function with respect to their immunoregulatory capacity.
Collapse
Affiliation(s)
- Sibel Kara
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jennifer J Lühr
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Division of Nano-Optics, Max-Planck Institute for the Science of Light, Erlangen, Germany
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Anja Lux
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
7
|
Wist M, Meier L, Gutman O, Haas J, Endres S, Zhou Y, Rösler R, Wiese S, Stilgenbauer S, Hobeika E, Henis YI, Gierschik P, Walliser C. Noncatalytic Bruton's tyrosine kinase activates PLCγ 2 variants mediating ibrutinib resistance in human chronic lymphocytic leukemia cells. J Biol Chem 2020; 295:5717-5736. [PMID: 32184360 DOI: 10.1074/jbc.ra119.011946] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/26/2020] [Indexed: 12/25/2022] Open
Abstract
Treatment of patients with chronic lymphocytic leukemia (CLL) with inhibitors of Bruton's tyrosine kinase (BTK), such as ibrutinib, is limited by primary or secondary resistance to this drug. Examinations of CLL patients with late relapses while on ibrutinib, which inhibits BTK's catalytic activity, revealed several mutations in BTK, most frequently resulting in the C481S substitution, and disclosed many mutations in PLCG2, encoding phospholipase C-γ2 (PLCγ2). The PLCγ2 variants typically do not exhibit constitutive activity in cell-free systems, leading to the suggestion that in intact cells they are hypersensitive to Rac family small GTPases or to the upstream kinases spleen-associated tyrosine kinase (SYK) and Lck/Yes-related novel tyrosine kinase (LYN). The sensitivity of the PLCγ2 variants to BTK itself has remained unknown. Here, using genetically-modified DT40 B lymphocytes, along with various biochemical assays, including analysis of PLCγ2-mediated inositol phosphate formation, inositol phospholipid assessments, fluorescence recovery after photobleaching (FRAP) static laser microscopy, and determination of intracellular calcium ([Ca2+] i ), we show that various CLL-specific PLCγ2 variants such as PLCγ2S707Y are hyper-responsive to activated BTK, even in the absence of BTK's catalytic activity and independently of enhanced PLCγ2 phospholipid substrate supply. At high levels of B-cell receptor (BCR) activation, which may occur in individual CLL patients, catalytically-inactive BTK restored the ability of the BCR to mediate increases in [Ca2+] i Because catalytically-inactive BTK is insensitive to active-site BTK inhibitors, the mechanism involving the noncatalytic BTK uncovered here may contribute to preexisting reduced sensitivity or even primary resistance of CLL to these drugs.
Collapse
Affiliation(s)
- Martin Wist
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Laura Meier
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Orit Gutman
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Jennifer Haas
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Sascha Endres
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Yuan Zhou
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Reinhild Rösler
- Core Unit Mass Spectrometry and Proteomics, Medical Faculty, Ulm University Medical Center, 89081 Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Medical Faculty, Ulm University Medical Center, 89081 Ulm, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Elias Hobeika
- Institute of Immunology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Peter Gierschik
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany.
| | - Claudia Walliser
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany.
| |
Collapse
|
8
|
Ben-Shmuel A, Biber G, Barda-Saad M. Unleashing Natural Killer Cells in the Tumor Microenvironment-The Next Generation of Immunotherapy? Front Immunol 2020; 11:275. [PMID: 32153582 PMCID: PMC7046808 DOI: 10.3389/fimmu.2020.00275] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence of immunotherapy for cancer treatment bears considerable clinical promise. Nevertheless, many patients remain unresponsive, acquire resistance, or suffer dose-limiting toxicities. Immune-editing of tumors assists their escape from the immune system, and the tumor microenvironment (TME) induces immune suppression through multiple mechanisms. Immunotherapy aims to bolster the activity of immune cells against cancer by targeting these suppressive immunomodulatory processes. Natural Killer (NK) cells are a heterogeneous subset of immune cells, which express a diverse array of activating and inhibitory germline-encoded receptors, and are thus capable of directly targeting and killing cancer cells without the need for MHC specificity. Furthermore, they play a critical role in triggering the adaptive immune response. Enhancing the function of NK cells in the context of cancer is therefore a promising avenue for immunotherapy. Different NK-based therapies have been evaluated in clinical trials, and some have demonstrated clinical benefits, especially in the context of hematological malignancies. Solid tumors remain much more difficult to treat, and the time point and means of intervention of current NK-based treatments still require optimization to achieve long term effects. Here, we review recently described mechanisms of cancer evasion from NK cell immune surveillance, and the therapeutic approaches that aim to potentiate NK function. Specific focus is placed on the use of specialized monoclonal antibodies against moieties on the cancer cell, or on both the tumor and the NK cell. In addition, we highlight newly identified mechanisms that inhibit NK cell activity in the TME, and describe how biochemical modifications of the TME can synergize with current treatments and increase susceptibility to NK cell activity.
Collapse
Affiliation(s)
- Aviad Ben-Shmuel
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Guy Biber
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Mira Barda-Saad
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
9
|
Sanseviero E. NK Cell-Fc Receptors Advance Tumor Immunotherapy. J Clin Med 2019; 8:E1667. [PMID: 31614774 PMCID: PMC6832859 DOI: 10.3390/jcm8101667] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy has revolutionized the treatment of cancer patients. Among immunotherapeutic approaches, antibodies targeting immune checkpoint inhibitors Programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are approved for treatment of metastatic melanoma and are in clinical trials for a variety of other cancers. The contribution of Natural Killer (NK) cells to the efficacy of immune checkpoint inhibitors is becoming more evident. Enhancing both T and NK cell function in cancer could result in a robust and durable response. Along with the ability to directly kill tumor cells, NK cells can mediate antibody-dependent cellular cytotoxicity (ADCC) given the expression of Fragment Crystallizable (Fc) receptors. Promising novel antibodies modified with improved Fc-receptor-mediated functions or Fc-engagers to kill target cells have been tested in pre-clinical models with considerable results. Combination therapies with immune-therapeutic antibodies with enhancers of NK-cell Fc-receptor-mediated function can be exploited to increase the efficacy of these antibodies. Herein, I discuss possible strategies to improve the success of immunotherapy by boosting NK cell function.
Collapse
Affiliation(s)
- Emilio Sanseviero
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Dominant role of splenic marginal zone lipid rafts in the classical complement pathway against S. pneumoniae. Cell Death Discov 2019; 5:133. [PMID: 31531231 PMCID: PMC6733876 DOI: 10.1038/s41420-019-0213-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/11/2019] [Accepted: 08/18/2019] [Indexed: 12/28/2022] Open
Abstract
Lipid rafts (LRs) play crucial roles in complex physiological processes, modulating innate and acquired immune responses to pathogens. The transmembrane C-type lectins human dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) and its mouse homolog SIGN-R1 are distributed in LRs and expressed on splenic marginal zone (MZ) macrophages. The DC-SIGN-C1q or SIGN-R1-C1q complex could mediate the immunoglobulin (Ig)-independent classical complement pathway against Streptococcus pneumoniae. Precise roles of LRs during this complement pathway are unknown. Here we show that LRs are indispensable for accelerating the DC-SIGN- or SIGN-R1-mediated classical complement pathway against S. pneumoniae, thus facilitating rapid clearance of the pathogen. The trimolecular complex of SIGN-R1-C1q-C4 was exclusively enriched in LRs of splenic MZ macrophages and their localization was essential for activating C3 catabolism and enhancing pneumococcal clearance, which were abolished in SIGN-R1-knockout mice. However, DC-SIGN replacement on splenic MZ macrophage’s LRs of SIGN-R1-depleted mice reversed these defects. Disruption of LRs dramatically reduced pneumococcal uptake and decomposition. Additionally, DC- SIGN, C1q, C4, and C3 were obviously distributed in splenic LRs of cadavers. Therefore, LRs on splenic SIGN-R1+ or DC-SIGN+ macrophages could provide spatially confined and optimal bidirectional platforms, not only for usual intracellular events, for example recognition and phagocytosis of pathogens, but also an unusual extracellular event such as the complement system. These findings improve our understanding of the orchestrated roles of the spleen, unraveling a new innate immune system initiated from splenic MZ LRs, and yielding answers to several long-standing problems, including the need to understand the profound role of LRs in innate immunity, the need to identify how such a small portion of splenic SIGN-R1+ macrophages (<0.05% of splenic macrophages) effectively resist S. pneumoniae, and the need to understand how LRs can promote the protective function of DC-SIGN against S. pneumoniae in the human spleen.
Collapse
|
11
|
Pinette A, McMichael E, Courtney NB, Duggan M, Benner BN, Choueiry F, Yu L, Abood D, Mace TA, Carson WE. An IL-15-based superagonist ALT-803 enhances the NK cell response to cetuximab-treated squamous cell carcinoma of the head and neck. Cancer Immunol Immunother 2019; 68:1379-1389. [PMID: 31338557 PMCID: PMC7032639 DOI: 10.1007/s00262-019-02372-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 07/16/2019] [Indexed: 01/17/2023]
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) is the sixth most common cancer worldwide and epidermal growth factor receptor (EGFR) is overexpressed in greater than 90% of patient tumors. Cetuximab is a monoclonal antibody that binds to EGFR and can activate immune cells, such as natural killer (NK) cells, that express receptors for the Fc (constant region) of immunoglobulin G. IL-15 (interleukin-15) is a critical factor for the development, proliferation and activation of effector NK cells. A novel IL-15 compound known as ALT-803 that consists of genetically modified IL-15 plus the IL-15 receptor alpha protein (IL15Rα) fused to the Fc portion of IgG1 has recently been developed. We hypothesized that treatment with ALT-803 would increase NK cell-mediated cytotoxicity of cetuximab-coated head and neck squamous cells. CD56+ NK cells from normal healthy donors were treated overnight with ALT-803 and tested for their ability to lyse cetuximab-coated tumor cells. Cytotoxicity was greater following NK cell ALT-803 activation, as compared to controls. ALT-803-treated NK cells secreted significantly higher levels of IFN-γ than control conditions. Additionally, NK cells showed increased levels of phospho-ERK and phospho-STAT5 when co-cultured with cetuximab-coated tumors and ALT-803. Administration of both cetuximab and ALT-803 to mice harboring Cal27 SCCHN tumors resulted in significantly decreased tumor volume when compared to controls and compared to single-agent treatment alone. Overall, the present data suggest that cetuximab treatment in combination with ALT-803 in patients with EGFR-positive SCCHN may result in significant NK cell activation and have important anti-tumor activity.
Collapse
Affiliation(s)
- Ashley Pinette
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Elizabeth McMichael
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | | | - Megan Duggan
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Brooke N Benner
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Fouad Choueiry
- Division of Gastroenterology, Hepatology & Nutrition, Department of Internal Medicine, The Ohio State University, 420 W 12th Ave., Columbus, OH, 43210, USA
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - David Abood
- Division of Gastroenterology, Hepatology & Nutrition, Department of Internal Medicine, The Ohio State University, 420 W 12th Ave., Columbus, OH, 43210, USA
| | - Thomas A Mace
- Division of Gastroenterology, Hepatology & Nutrition, Department of Internal Medicine, The Ohio State University, 420 W 12th Ave., Columbus, OH, 43210, USA.
| | - William E Carson
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
12
|
McMichael EL, Benner B, Atwal LS, Courtney NB, Mo X, Davis ME, Campbell AR, Duggan MC, Williams K, Martin K, Levine K, Olaverria Salavaggione GN, Noel T, Ganju A, Uppati S, Paul B, Olencki T, Teknos TN, Savvides P, Tridandapani S, Byrd JC, Caligiuri MA, Liu SV, Carson WE. A Phase I/II Trial of Cetuximab in Combination with Interleukin-12 Administered to Patients with Unresectable Primary or Recurrent Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2019; 25:4955-4965. [PMID: 31142501 DOI: 10.1158/1078-0432.ccr-18-2108] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 03/05/2019] [Accepted: 05/10/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE mAbs including cetuximab can induce antibody-dependent cellular cytotoxicity (ADCC) and cytokine production mediated via innate immune cells with the ability to recognize mAb-coated tumors. Preclinical modeling has shown that costimulation of natural killer (NK) cells via the Fc receptor and the IL12 receptor promotes NK-cell-mediated ADCC and production of cytokines. PATIENTS AND METHODS This phase I/II trial evaluated the combination of cetuximab with IL12 for the treatment of EGFR-expressing head and neck cancer. Treatment consisted of cetuximab 500 mg/m2 i.v. every 2 weeks with either 0.2 mcg/kg or 0.3 mcg/kg IL12 s.c. on days 2 and 5 of the 2-week cycle, beginning with cycle 2. Correlative studies from blood draws obtained prior to treatment and during therapy included measurement of ADCC, serum cytokine, and chemokine analysis, determination of NK cell FcγRIIIa polymorphisms, and an analysis of myeloid-derived suppressor cell (MDSC) frequency in peripheral blood. RESULTS The combination of cetuximab and IL12 was well tolerated. No clinical responses were observed, however, 48% of patients exhibited prolonged progression-free survival (PFS; average of 6.5 months). Compared with patients that did not exhibit clinical benefit, patients with PFS >100 days exhibited increased ADCC as therapy continued compared with baseline, greater production of IFNγ, IP-10, and TNFα at the beginning of cycle 8 compared with baseline values and had a predominance of monocytic MDSCs versus granulocytic MDSCs prior to therapy. CONCLUSIONS Further investigation of IL12 as an immunomodulatory agent in combination with cetuximab in head and neck squamous cell carcinoma is warranted.
Collapse
Affiliation(s)
- Elizabeth L McMichael
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Brooke Benner
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Lakhvir S Atwal
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | | | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Melanie E Davis
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Amanda R Campbell
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Megan C Duggan
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Kallan Williams
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Kyle Martin
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Kala Levine
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | | | - Tiffany Noel
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Akaansha Ganju
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Sarvani Uppati
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Bonnie Paul
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Thomas Olencki
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | | | - Panos Savvides
- Department of Medicine, Division of Hematology/Oncology, The Ohio State University, Columbus, Ohio
| | | | - John C Byrd
- Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Michael A Caligiuri
- Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Stephen V Liu
- Department of Medicine, Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, D.C
| | - William E Carson
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio. .,Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
13
|
Benner B, Scarberry L, Suarez-Kelly LP, Duggan MC, Campbell AR, Smith E, Lapurga G, Jiang K, Butchar JP, Tridandapani S, Howard JH, Baiocchi RA, Mace TA, Carson WE. Generation of monocyte-derived tumor-associated macrophages using tumor-conditioned media provides a novel method to study tumor-associated macrophages in vitro. J Immunother Cancer 2019; 7:140. [PMID: 31138333 PMCID: PMC6540573 DOI: 10.1186/s40425-019-0622-0] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/16/2019] [Indexed: 02/08/2023] Open
Abstract
Background Tumor-associated macrophages (TAM) are expanded and exhibit tumor-promoting properties within the tumor microenvironment. Current methods to study TAM have not been replicated across cancer types and often do not include exogenous growth factors from the tumor, a key factor in TAM differentiation in vivo. Methods In this study, an in vitro method to generate monocyte- derived TAM using tumor- conditioned media (TCM) and a cytokine cocktail containing IL-4, IL-10, and M-CSF was utilized to study the phenotype, morphology, and function of TAM across multiple cancer types. TCM was generated from two breast cancer cell lines and an Epstein-Barr virus-positive lymphoma cell line. The properties of in vitro generated TAM were compared to in vitro generated M1 and M2- like macrophages and TAM isolated from patients with cancer. Results TAM generated in this fashion displayed an increase in CD163/CD206 co-expression compared to M2- like macrophages (87 and 36%, respectively). TAM generated in vitro exhibited increased transcript levels of the functional markers IL-6, IL-10, CCL2, c-Myc, iNOS, and arginase compared to in vitro generated M2-like macrophages. Functionally, in vitro generated TAM inhibited the proliferation of T cells (47% decrease from M1-like macrophages) and the production of IFN-γ by natural killer cells was inhibited (44%) when co-cultured with TAM. Furthermore, in vitro generated TAM secreted soluble factors that promote the growth and survival of tumor cells. Conclusions Limited access to patient TAM highlights the need for methods to generate TAM in vitro. Our data confirm that monocyte-derived TAM can be generated reliably using TCM plus the cytokine cocktail of IL-4, IL-10, and M-CSF. Given the ability of TAM to inhibit immune cell function, continued study of methods to deplete or deactivate TAM in the setting of cancer are warranted. Electronic supplementary material The online version of this article (10.1186/s40425-019-0622-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Brooke Benner
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Luke Scarberry
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Lorena P Suarez-Kelly
- Division of Surgical Oncology, The Ohio State University, N924 Doan Hall, 410 W. 10th Avenue, Columbus, OH, 43210, USA
| | - Megan C Duggan
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Amanda R Campbell
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Emily Smith
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Gabriella Lapurga
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kallie Jiang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Jonathan P Butchar
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | | | - John Harrison Howard
- Division of Surgical Oncology, The Ohio State University, N924 Doan Hall, 410 W. 10th Avenue, Columbus, OH, 43210, USA
| | - Robert A Baiocchi
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, OH, USA
| | - Thomas A Mace
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA. .,Division of Surgical Oncology, The Ohio State University, N924 Doan Hall, 410 W. 10th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
14
|
CD33 (Siglec-3) Inhibitory Function: Role in the NKG2D/DAP10 Activating Pathway. J Immunol Res 2019; 2019:6032141. [PMID: 31143782 PMCID: PMC6501159 DOI: 10.1155/2019/6032141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/25/2018] [Accepted: 02/28/2019] [Indexed: 12/14/2022] Open
Abstract
CD33 (siglec-3), a well-known target in leukemia therapy, is an inhibitory sialoadhesin expressed in human leukocytes of the myeloid lineage and some lymphoid subsets, including NK cells. It may constitute a control mechanism of the innate immune system; nevertheless, its role as an inhibitory receptor remains elusive. Using human NK cells as a cellular model, we analyzed CD33 inhibitory function upon different activating receptors. In high-cytotoxicity NKL cells, CD33 displayed a prominent inhibition on cytotoxicity triggered by the activating receptors NKG2D and, in a lower extent, 2B4, whereas it did not inhibit NKp46-induced cytotoxicity. NKp46 was partially inhibited by CD33 only when low-cytotoxicity NKL cells were tested. CD33 triggering did not inhibit IFN-γ secretion, contrasting with ILT-2 and CD94/NKG2A inhibitory receptors that inhibited cytotoxicity and IFN-γ secretion induced by all activating receptors tested. CD33-mediated inhibition of NKG2D-induced triggering involved Vav1 dephosphorylation. Our results support the role of CD33 as an inhibitory receptor preferentially regulating the NKG2D/DAP10 cytotoxic signaling pathway, which could be involved in self-tolerance and tumor and infected cell recognition.
Collapse
|
15
|
Pahl JHW, Cerwenka A, Ni J. Memory-Like NK Cells: Remembering a Previous Activation by Cytokines and NK Cell Receptors. Front Immunol 2018; 9:2796. [PMID: 30546366 PMCID: PMC6279934 DOI: 10.3389/fimmu.2018.02796] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/13/2018] [Indexed: 12/24/2022] Open
Abstract
Natural Killer (NK) cells are cytotoxic innate lymphoid cells serving at the front line against infection and cancer. In inflammatory microenvironments, multiple soluble and contact-dependent signals modulate NK cell responsiveness. Besides their innate cytotoxic and immunostimulatory activity, it has been uncovered in recent years that NK cells constitute a heterogeneous and versatile cell subset. Persistent memory-like NK populations that mount a robust recall response were reported during viral infection, contact hypersensitivity reactions, and after stimulation by pro-inflammatory cytokines or activating receptor pathways. In this review, we highlight recent findings on the generation, functionality, and clinical applicability of memory-like NK cells and describe common features in comparison to other recent concepts of memory NK cells. Understanding of these features will facilitate the conception and design of novel NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Jens H W Pahl
- Department for Immunobiochemistry, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany
| | - Adelheid Cerwenka
- Department for Immunobiochemistry, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany
| | - Jing Ni
- Department for Immunobiochemistry, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany.,Innate Immunity, German Cancer Consortium, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
16
|
Liu Z, Yang W, Yang S, Cai K. The close association between IL‑12Rβ2 and p38MAPK, and higher expression in the early stages of NSCLC, indicates a good prognosis for survival. Mol Med Rep 2018; 18:2307-2313. [PMID: 29956791 DOI: 10.3892/mmr.2018.9206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 04/16/2018] [Indexed: 11/05/2022] Open
Abstract
Interleukin‑12 receptor (IL‑12R) and p38 mitogen‑activated protein kinase (p38MAPK) serve an important role in non‑small cell lung cancer (NSCLC). It has previously been suggested that IL‑12Rβ2 may be involved in key regulatory pathways and interacts with the p38MAPK signaling pathway. The present study aimed to elucidate the possible association and roles of IL‑12Rβ2 and p38MAPK in NSCLC. The protein expression levels of IL‑12Rβ2 and p38MAPK were measured in 230 NSCLC tissue samples by immunohistochemistry (IHC) and western blot analyses. In addition, an immunofluorescence assay was used to observe the expression levels of these proteins in A549 and H358 cells. The associations between IL‑12Rβ2, p38MAPK and clinical characteristics, were evaluated by Pearson χ2 and Spearman correlation tests. Kaplan‑Meier plots (log‑rank test) and Cox proportional hazard models were used to analyze overall survival (OS). Compared with in benign pulmonary tissues, the expression levels of IL‑12Rβ2 and p38MAPK were not demonstrated to be significantly different in I+II pathological tumor‑node‑metastasis (pTNM) stage NSCLC tissues; however, reduced expression was detected in III+IV pTNM stage NSCLC tissues. Analysis of the association between advanced stage pTNM and the expression of both proteins demonstrated a significantly decreased Allred score (both P<0.0001), which was confirmed by IHC and western blot analyses. The IHC results demonstrated a significant correlation between IL‑12Rβ2 and p38MAPK expression (r=0.415, P=0.0143). By analyzing IL‑12Rβ2, p38MAPK expression and clinical characteristics, it was identified that IL‑12Rβ2 was significantly associated with gender (P=0.0168), age (P=0.0341), histological type (P<0.0001) and pTNM stage (P<0.0001). p38MAPK demonstrated a strong association with gender (P=0.0082) and pTNM stage (P<0.0001). The results of a Kaplan‑Meier analysis indicated that positive IL‑12Rβ2 and p38MAPK expression was associated with increased OS compared with negative protein expression. The Cox proportional hazard models revealed that IL‑12Rβ2 and p38MAPK predicted a long OS. To the best of our knowledge, the present study is the first to reveal a close association between IL‑12Rβ2 and p38MAPK, and their possible function in NSCLC progression. It further demonstrated that expression of both proteins was lower with advanced pTNM staging, whereas a high expression of both proteins was associated with improved prognosis in NSCLC.
Collapse
Affiliation(s)
- Zhaoguo Liu
- Department of General Thoracic Surgery, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510089, P.R. China
| | - Weilin Yang
- Department of General Thoracic Surgery, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510089, P.R. China
| | - Shibin Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510089, P.R. China
| | - Kaican Cai
- Department of Cardio‑Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510089, P.R. China
| |
Collapse
|
17
|
Sasaki K, Miyashita Y, Asai D, Funamoto D, Sato K, Yamaguchi Y, Mishima Y, Iino T, Takaishi S, Nagano J, Kishimura A, Mori T, Katayama Y. A peptide inhibitor of antibody-dependent cell-mediated cytotoxicity against EGFR/folate receptor-α double positive cells. MEDCHEMCOMM 2018; 9:783-788. [PMID: 30108967 PMCID: PMC6072457 DOI: 10.1039/c8md00010g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/24/2018] [Indexed: 12/20/2022]
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) is caused by natural killer (NK) cells upon recognition of antigen-bound IgG via FcγRIIIa. This mechanism is crucial for cytolysis of pathogen-infected cells and monoclonal antibody (mAb)-mediated elimination of cancer cells. However, there is concern that mAb-based cancer therapy induces ADCC against non-target cells expressing antigens. To date, no strategy has been reported to enhance the selectivity of ADCC to protect non-target cells expressing antigens. Here, we introduce a model inhibitor which specifically blocks ADCC of anti-EGFR mAbs towards EGFR/folate receptor α (FRα) double positive cells. This inhibitor recruits mAbs on the FRα of the cell surface independent of Fab antigen recognition. The resulting ternary and/or quaternary complexes formed on the cell surface suppress signal transduction of FcγRIIIa in NK cells, consequently leading to more specific ADCC.
Collapse
Affiliation(s)
- Koichi Sasaki
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Yoshiki Miyashita
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Daisuke Asai
- Department of Microbiology , St. Marianna University School of Medicine , 2-16-1 Sugao, Miyamae-ku , Kawasaki , 216-8511 , Japan
| | - Daiki Funamoto
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Kazuki Sato
- Department of Environmental Science , Fukuoka Women's University , 1-1-1 Kasumigaoka , Fukuoka , 813-8529 , Japan
| | - Yoko Yamaguchi
- Department of Environmental Science , Fukuoka Women's University , 1-1-1 Kasumigaoka , Fukuoka , 813-8529 , Japan
| | - Yuji Mishima
- Clinical Chemotherapy , Cancer Chemotherapy Center , Japanese Foundation for Cancer Research , 3-8-31, Ariake, Koto-ku , Tokyo , 135-8550 , Japan
| | - Tadafumi Iino
- Centre for Advanced Medicine Innovation , Kyushu University , 3-1-1 Maidashi, Higashi-ku , Fukuoka , 812-8582 , Japan
| | - Shigeo Takaishi
- Centre for Advanced Medicine Innovation , Kyushu University , 3-1-1 Maidashi, Higashi-ku , Fukuoka , 812-8582 , Japan
| | - Jun Nagano
- Faculty of Arts and Science , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka 819-0395 , Fukuoka , Japan
| | - Akihiro Kishimura
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Takeshi Mori
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Yoshiki Katayama
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| |
Collapse
|
18
|
Pahl JHW, Koch J, Götz JJ, Arnold A, Reusch U, Gantke T, Rajkovic E, Treder M, Cerwenka A. CD16A Activation of NK Cells Promotes NK Cell Proliferation and Memory-Like Cytotoxicity against Cancer Cells. Cancer Immunol Res 2018. [PMID: 29514797 DOI: 10.1158/2326-6066.cir-17-0550] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD16A is a potent cytotoxicity receptor on human natural killer (NK) cells, which can be exploited by therapeutic bispecific antibodies. So far, the effects of CD16A-mediated activation on NK cell effector functions beyond classical antibody-dependent cytotoxicity have remained poorly elucidated. Here, we investigated NK cell responses after exposure to therapeutic antibodies such as the tetravalent bispecific antibody AFM13 (CD30/CD16A), designed for the treatment of Hodgkin lymphoma and other CD30+ lymphomas. Our results reveal that CD16A engagement enhanced subsequent IL2- and IL15-driven NK cell proliferation and expansion. This effect involved the upregulation of CD25 (IL2Rα) and CD132 (γc) on NK cells, resulting in increased sensitivity to low-dose IL2 or to IL15. CD16A engagement initially induced NK cell cytotoxicity. The lower NK cell reactivity observed 1 day after CD16A engagement could be recovered by reculture in IL2 or IL15. After reculture in IL2 or IL15, these CD16A-experienced NK cells exerted more vigorous IFNγ production upon restimulation with tumor cells or cytokines. Importantly, after reculture, CD16A-experienced NK cells also exerted increased cytotoxicity toward different tumor targets, mainly through the activating NK cell receptor NKG2D. Our findings uncover a role for CD16A engagement in priming NK cell responses to restimulation by cytokines and tumor cells, indicative of a memory-like functionality. Our study suggests that combination of AFM13 with IL2 or IL15 may boost NK cell antitumor activity in patients by expanding tumor-reactive NK cells and enhancing NK cell reactivity, even upon repeated tumor encounters. Cancer Immunol Res; 6(5); 517-27. ©2018 AACR.
Collapse
Affiliation(s)
- Jens H W Pahl
- Innate Immunity, German Cancer Research Center, Heidelberg, Germany.,Department for Immunobiochemistry, Medical Faculty Mannheim, Heidelberg University, Germany
| | | | - Jana-Julia Götz
- Innate Immunity, German Cancer Research Center, Heidelberg, Germany
| | - Annette Arnold
- Innate Immunity, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | - Adelheid Cerwenka
- Innate Immunity, German Cancer Research Center, Heidelberg, Germany. .,Department for Immunobiochemistry, Medical Faculty Mannheim, Heidelberg University, Germany
| |
Collapse
|
19
|
Stiff A, Trikha P, Mundy-Bosse B, McMichael E, Mace TA, Benner B, Kendra K, Campbell A, Gautam S, Abood D, Landi I, Hsu V, Duggan M, Wesolowski R, Old M, Howard JH, Yu L, Stasik N, Olencki T, Muthusamy N, Tridandapani S, Byrd JC, Caligiuri M, Carson WE. Nitric Oxide Production by Myeloid-Derived Suppressor Cells Plays a Role in Impairing Fc Receptor-Mediated Natural Killer Cell Function. Clin Cancer Res 2018; 24:1891-1904. [PMID: 29363526 DOI: 10.1158/1078-0432.ccr-17-0691] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 11/22/2017] [Accepted: 01/19/2018] [Indexed: 12/14/2022]
Abstract
Purpose: mAbs are used to treat solid and hematologic malignancies and work in part through Fc receptors (FcRs) on natural killer cells (NK). However, FcR-mediated functions of NK cells from patients with cancer are significantly impaired. Identifying the mechanisms of this dysfunction and impaired response to mAb therapy could lead to combination therapies and enhance mAb therapy.Experimental Design: Cocultures of autologous NK cells and MDSC from patients with cancer were used to study the effect of myeloid-derived suppressor cells (MDSCs) on NK-cell FcR-mediated functions including antibody-dependent cellular cytotoxicity, cytokine production, and signal transduction in vitro Mouse breast cancer models were utilized to study the effect of MDSCs on antibody therapy in vivo and test the efficacy of combination therapies including a mAb and an MDSC-targeting agent.Results: MDSCs from patients with cancer were found to significantly inhibit NK-cell FcR-mediated functions including antibody-dependent cellular cytotoxicity, cytokine production, and signal transduction in a contact-independent manner. In addition, adoptive transfer of MDSCs abolished the efficacy of mAb therapy in a mouse model of pancreatic cancer. Inhibition of iNOS restored NK-cell functions and signal transduction. Finally, nonspecific elimination of MDSCs or inhibition of iNOS in vivo significantly improved the efficacy of mAb therapy in a mouse model of breast cancer.Conclusions: MDSCs antagonize NK-cell FcR-mediated function and signal transduction leading to impaired response to mAb therapy in part through nitric oxide production. Thus, elimination of MDSCs or inhibition of nitric oxide production offers a strategy to improve mAb therapy. Clin Cancer Res; 24(8); 1891-904. ©2018 AACR.
Collapse
Affiliation(s)
- Andrew Stiff
- Medical Scientist Training Program, Columbus, Ohio.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Prashant Trikha
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | | | - Elizabeth McMichael
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Thomas A Mace
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Brooke Benner
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Kari Kendra
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Amanda Campbell
- Medical Scientist Training Program, Columbus, Ohio.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Shalini Gautam
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - David Abood
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Ian Landi
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Vincent Hsu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Megan Duggan
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Matthew Old
- Department of Otolaryngology, The Ohio State University, Columbus, Ohio
| | - John Harrison Howard
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Nancy Stasik
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Thomas Olencki
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Natarajan Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Susheela Tridandapani
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine and Dorothy M. Davis Heart and Lung Research Institute, Columbus, Ohio
| | - John C Byrd
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Michael Caligiuri
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio. .,Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio
| |
Collapse
|
20
|
Ziblat A, Nuñez SY, Raffo Iraolagoitia XL, Spallanzani RG, Torres NI, Sierra JM, Secchiari F, Domaica CI, Fuertes MB, Zwirner NW. Interleukin (IL)-23 Stimulates IFN-γ Secretion by CD56 bright Natural Killer Cells and Enhances IL-18-Driven Dendritic Cells Activation. Front Immunol 2018; 8:1959. [PMID: 29403472 PMCID: PMC5785728 DOI: 10.3389/fimmu.2017.01959] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 12/19/2017] [Indexed: 12/29/2022] Open
Abstract
Interleukin (IL)-23 is a member of the IL-12 family of cytokines that, as the other members of this family, is secreted by monocytes, macrophages, and dendritic cells (DC) upon recognition of bacterial, viral, and fungal components. IL-23 is critical during immunity against acute infections, and it is also involved in the development of autoimmune diseases. Although immunoregulatory effects of IL-23 on mouse natural killer (NK) cells have been described, the effect of IL-23 on human NK cells remains ill-defined. In this study, we observed that monocytes stimulated with LPS secreted IL-23 and that blockade of this cytokine during monocyte and NK cell coculture led to a diminished production of IFN-γ by NK cells. Accordingly, rIL-23-induced NK cell activation and stimulated IFN-γ production by CD56bright NK cells. This effect involved MEK1/MEK2, JNK, PI3K, mammalian target of rapamycin, and NF-κB, but not STAT-1, STAT-3, nor p38 MAPK pathways. Moreover, while NK cell-mediated cytotoxicity remained unaltered, antibody-dependent cellular cytotoxicity (ADCC) was enhanced after IL-23 stimulation. In addition, IL-23 displayed a synergistic effect with IL-18 for IFN-γ production by both CD56bright and CD56dim NK cells, and this effect was due to a priming effect of IL-23 for IL-18 responsiveness. Furthermore, NK cells pre-stimulated with IL-18 promoted an increase in CD86 expression and IL-12 secretion by DC treated with LPS, and IL-23 potentiated these effects. Moreover, IL-23-driven enhancement of NK cell “helper” function was dependent on NK cell-derived IFN-γ. Therefore, our results suggest that IL-23 may trigger NK cell-mediated “helper” effects on adaptive immunity, shaping T cell responses during different pathological situations through the regulation of DC maturation.
Collapse
Affiliation(s)
- Andrea Ziblat
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Sol Y Nuñez
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Ximena Lucía Raffo Iraolagoitia
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Raúl German Spallanzani
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Nicolás I Torres
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Jessica M Sierra
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Florencia Secchiari
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Carolina I Domaica
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Mercedes B Fuertes
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina
| | - Norberto W Zwirner
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Laboratorio de Fisiopatología de la Inmunidad Innata, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
21
|
Duggan MC, Campbell AR, McMichael EL, Opheim KS, Levine KM, Bhave N, Culbertson MC, Noel T, Yu L, Carson WE. Co-stimulation of the fc receptor and interleukin-12 receptor on human natural killer cells leads to increased expression of cd25. Oncoimmunology 2017; 7:e1381813. [PMID: 29308301 DOI: 10.1080/2162402x.2017.1381813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/31/2017] [Accepted: 09/14/2017] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells serve a critical role in the immune response against microbes and developing tumors. We have demonstrated that NK cells produce stimulatory cytokines (e.g., IFN-γ) in response to potent stimulation via immobilized IgG (to engage Fc receptors) and interleukin (IL)-12. CD25 is a component of the high-affinity IL-2R, which promotes NK cell activation in response to low doses of IL-2 such as those released by activated T cells. We hypothesized that stimulation of NK cells via IgG and IL-12 would enhance CD25 expression and promote NK cell anti-tumor activity in response to low-dose IL-2. It was confirmed that this dual stimulation strategy significantly enhanced NK cell CD25 expression compared to unstimulated cells or cells treated with IgG or IL-12 alone. Dual stimulated NK cells also were more responsive to low-dose IL-2. Dual stimulated NK cells subsequently treated with low-dose IL-2 (10 pg/mL) displayed enhanced intracellular signaling as indicated by increased pSTAT5 levels. IFN-γ production and cytotoxicity against K562 cells by NK cells stimulated with low-dose IL-2 was comparable to that of cells treated with high-dose IL-2 (10 ng/mL). Importantly, cells isolated from head and neck cancer patients receiving the mAb cetuximab and IL-12 on a clinical trial displayed increased CD25 expression following combination therapy compared to baseline. Altogether, these findings suggest that FcR and IL-12R co-stimulation induces expression of the high-affinity IL-2R and promotes NK cell anti-tumor activity.
Collapse
Affiliation(s)
- Megan C Duggan
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Amanda R Campbell
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH.,Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH
| | - Elizabeth L McMichael
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Kallan S Opheim
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Kala M Levine
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Neela Bhave
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Michelle C Culbertson
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Tiffany Noel
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University, Columbus, OH
| | - W E Carson
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH.,Department of Surgery, The Ohio State University, Columbus, OH
| |
Collapse
|
22
|
Campbell AR, Duggan MC, Suarez-Kelly LP, Bhave N, Opheim KS, McMichael EL, Trikha P, Parihar R, Luedke E, Lewis A, Yung B, Lee R, Raulet D, Tridandapani S, Groh V, Yu L, Yildiz V, Byrd JC, Caligiuri MA, Carson WE. MICA-Expressing Monocytes Enhance Natural Killer Cell Fc Receptor-Mediated Antitumor Functions. Cancer Immunol Res 2017; 5:778-789. [PMID: 28724544 DOI: 10.1158/2326-6066.cir-16-0005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 07/20/2016] [Accepted: 07/05/2017] [Indexed: 12/11/2022]
Abstract
Natural killer (NK) cells are large granular lymphocytes that promote the antitumor response via communication with other cell types in the tumor microenvironment. Previously, we have shown that NK cells secrete a profile of immune stimulatory factors (e.g., IFNγ, MIP-1α, and TNFα) in response to dual stimulation with the combination of antibody (Ab)-coated tumor cells and cytokines, such as IL12. We now demonstrate that this response is enhanced in the presence of autologous monocytes. Monocyte enhancement of NK cell activity was dependent on cell-to-cell contact as determined by a Transwell assay. It was hypothesized that NK cell effector functions against Ab-coated tumor cells were enhanced via binding of MICA on monocytes to NK cell NKG2D receptors. Strategies to block MICA-NKG2D interactions resulted in reductions in IFNγ production. Depletion of monocytes in vivo resulted in decreased IFNγ production by murine NK cells upon exposure to Ab-coated tumor cells. In mice receiving trastuzumab and IL12 therapy, monocyte depletion resulted in significantly greater tumor growth in comparison to mock-depleted controls (P < 0.05). These data suggest that NK cell-monocyte interactions enhance NK cell antitumor activity in the setting of monoclonal Ab therapy for cancer. Cancer Immunol Res; 5(9); 778-89. ©2017 AACR.
Collapse
Affiliation(s)
- Amanda R Campbell
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Megan C Duggan
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | | | - Neela Bhave
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Kallan S Opheim
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Elizabeth L McMichael
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Prashant Trikha
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Robin Parihar
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Eric Luedke
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Adrian Lewis
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Bryant Yung
- Department of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Robert Lee
- Department of Pharmacy, The Ohio State University, Columbus, Ohio
| | - David Raulet
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - Susheela Tridandapani
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, The Ohio State University, Columbus, Ohio
| | - Veronika Groh
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Vedat Yildiz
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - John C Byrd
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Michael A Caligiuri
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Division of Hematology, The Ohio State University, Columbus, Ohio
| | - William E Carson
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio. .,Department of Surgery, The Ohio State University, Columbus, Ohio
| |
Collapse
|
23
|
McMichael EL, Courtney NB, Duggan MC, Wesolowski R, Quiroga D, Kondadasula SV, Atwal LS, Bhave N, Luedke E, Jaime-Ramirez AC, Campbell AR, Mo X, Byrd JC, Carson Iii WE. Activation of the FcgammaReceptorIIIa on human natural killer cells leads to increased expression of functional interleukin-21 receptor. Oncoimmunology 2017. [PMID: 28638738 DOI: 10.1080/2162402x.2017.1312045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Natural killer (NK) cells are innate immune effector cells that play a crucial role in immune surveillance and the destruction of cancer cells. NK cells express a low-affinity receptor for the Fc or constant region of immunoglobulin G (FcγRIIIa) and multiple cytokine receptors that respond to antibody-coated targets and cytokines in the tumor microenvironment. In the present work, microarray gene expression analysis revealed that the IL-21 receptor (IL-21R) was strongly upregulated following FcR stimulation. The IL-21R was found to be upregulated on FcR-stimulated NK cells at the transcript level as determined by reverse transcription polymerase chain reaction (RT-PCR). Immunoblot analysis revealed that protein expression of the IL-21R peaked at 8 h post-stimulation of the FcR. Inhibition of the mitogen-activated protein kinase (MAPK) pathway downstream of the FcR blocked the induction of IL-21R expression. Increased expression of the IL-21R sensitized NK cells to IL-21 stimulation, as treatment of FcR-stimulated NK cells led to significantly increased phosphorylation of STAT1 and STAT3, as measured by intracellular flow cytometry and immunoblot analysis. Following FcR-stimulation, IL-21-activated NK cells were better able to mediate the lysis of trastuzumab-coated human epidermal growth factor receptor 2 (HER2+) SK-BR-3 tumor cells as compared to control-treated cells. Likewise, IL-21-induced NK cell secretion of IFNγ following exposure to antibody-coated tumor cells was enhanced following FcR-stimulation. The analysis of NK cells from patients receiving trastuzumab therapy for HER2+ cancer exhibited increased levels of the IL-21R following the administration of antibody suggesting that the presence of monoclonal antibody-coated tumor cells in vivo can stimulate the increased expression of IL-21R on NK cells.
Collapse
Affiliation(s)
| | | | - Megan C Duggan
- Biomedical Sciences Graduate Program, College of Medicine, Columbus, OH, US
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, Columbus, OH, USA
| | - Dionisia Quiroga
- Division of Medical Oncology, Department of Internal Medicine, Columbus, OH, USA
| | | | | | - Neela Bhave
- Comprehensive Cancer Center, Columbus, OH, USA
| | - Eric Luedke
- Department of Surgery, Division of Surgical Oncology, Columbus, OH, USA
| | | | - Amanda R Campbell
- Biomedical Sciences Graduate Program, College of Medicine, Columbus, OH, US.,Medical Scientist Training Program, College of Medicine, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, Columbus, OH, USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, Columbus, OH, USA
| | - William E Carson Iii
- Department of Surgery, Division of Surgical Oncology, Columbus, OH, USA.,Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH, USA.,Biomedical Sciences Graduate Program, College of Medicine, Columbus, OH, US
| |
Collapse
|
24
|
Moy JD, Moskovitz JM, Ferris RL. Biological mechanisms of immune escape and implications for immunotherapy in head and neck squamous cell carcinoma. Eur J Cancer 2017; 76:152-166. [PMID: 28324750 DOI: 10.1016/j.ejca.2016.12.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/26/2016] [Accepted: 12/13/2016] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy with high morbidity and mortality. Despite advances in cytotoxic therapies and surgical techniques, overall survival (OS) has not improved over the past few decades. This emphasises the need for intense investigation into novel therapies with good tumour control and minimal toxicity. Cancer immunotherapy has led this endeavour, attempting to improve tumour recognition and expand immune responses against tumour cells. While various forms of HNSCC immunotherapy are in preclinical trials, the most promising direction thus far has been with monoclonal antibodies (mAbs), targeting growth factor and immune checkpoint receptors. Preclinical and early phase trials have shown unprecedented efficacy with minimal adverse effects. This article will review biological mechanisms of immune escape and implications for immunotherapy in HNSCC.
Collapse
Affiliation(s)
- Jennifer D Moy
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Fluorescent nanodiamonds engage innate immune effector cells: A potential vehicle for targeted anti-tumor immunotherapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:909-920. [PMID: 27993723 DOI: 10.1016/j.nano.2016.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/09/2016] [Accepted: 12/05/2016] [Indexed: 12/30/2022]
Abstract
Fluorescent nanodiamonds (FNDs) are nontoxic, infinitely photostable, and emit fluorescence in the near infrared region. Natural killer (NK) cells and monocytes are part of the innate immune system and are crucial to the control of carcinogenesis. FND-mediated stimulation of these cells may serve as a strategy to enhance anti-tumor activity. FNDs were fabricated with a diameter of 70±28 nm. Innate immune cell FND uptake, viability, surface marker expression, and cytokine production were evaluated in vitro. Evaluation of fluorescence emission from the FNDs was conducted in an animal model. In vitro results demonstrated that treatment of immune cells with FNDs resulted in significant dose-dependent FND uptake, no compromise in cell viability, and immune cell activation. FNDs were visualized in an animal model. Hence, FNDs may serve as novel agents with "track and trace" capabilities to stimulate innate immune cell anti-tumor responses, especially as FNDs are amenable to surface-conjugation with immunomodulatory molecules.
Collapse
|
26
|
Dasari S, Kathera C, Janardhan A, Praveen Kumar A, Viswanath B. Surfacing role of probiotics in cancer prophylaxis and therapy: A systematic review. Clin Nutr 2016; 36:1465-1472. [PMID: 27923508 DOI: 10.1016/j.clnu.2016.11.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/23/2016] [Accepted: 11/21/2016] [Indexed: 01/29/2023]
Abstract
Cancers figure among the most important causes of morbidity and mortality worldwide. Cancer and its associated infections are always complicated even when specific cancer regimens are available. It is well proved that Lactobacillus and other probiotic bacteria can modulate-ameliorate specific mechanisms against various infections including cancers. The present systematic review is intended to focus on the 'cellular and molecular mechanisms' of probiotic bacteria in the prevention and treatment of various cancers. The clinical and experimental findings of various studies explain the mechanisms such as apoptosis, antioxidant activity, immune response and epigenetics and illustrate the role of probiotics in cancer management and prophylaxis. In addition, the present review also discusses the safety aspects of probiotics when they are used in therapeutic and nutritional diet management. However, further investigations are required to reveal the effectiveness of probiotics in cancer treatment in clinical settings.
Collapse
Affiliation(s)
- Subramanyam Dasari
- Department of Biomedical Sciences, University of Illinois, College of Medicine at Rockford, Rockford, IL 61107, USA
| | - Chandrasekhar Kathera
- College of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing 210023, China
| | - Avilala Janardhan
- Department of Plant Biotechnology and Genomics, Centre for Biotechnology and Plant Genomics (CBGP), Polytechnic University of Madrid (UPM), Madrid 28040, Spain
| | - Arthala Praveen Kumar
- Department of Virology, College of Sciences, Sri Venkateswara University, Tirupati 517502, India
| | - Buddolla Viswanath
- Department of Bionanotechnology, Gachon University, San 65, Bokjeong dong, Sujeong gu, Seongnam si, Gyeonggi do 461 701, Republic of Korea.
| |
Collapse
|
27
|
Bertino EM, McMichael EL, Mo X, Trikha P, Davis M, Paul B, Grever M, Carson WE, Otterson GA. A Phase I Trial to Evaluate Antibody-Dependent Cellular Cytotoxicity of Cetuximab and Lenalidomide in Advanced Colorectal and Head and Neck Cancer. Mol Cancer Ther 2016; 15:2244-50. [PMID: 27458141 DOI: 10.1158/1535-7163.mct-15-0879] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 06/28/2016] [Indexed: 12/17/2022]
Abstract
mAbs can induce antibody-dependent cellular cytotoxicity (ADCC) via the innate immune system's ability to recognize mAb-coated cancer cells and activate immune effector cells. Lenalidomide is an immunomodulatory agent with the capacity to stimulate immune cell cytokine production and ADCC activity. This phase I trial evaluated the combination of cetuximab with lenalidomide for the treatment of advanced colorectal and head and neck squamous cell cancers (HNSCC). This trial included patients with advanced colorectal cancer or HNSCC. Treatment consisted of cetuximab 500 mg/m(2) i.v. every two weeks with lenalidomide given orally days 1-21 on a 28-day cycle. Three dose levels of lenalidomide were evaluated (15, 20, 25 mg). Correlative studies included measurement of ADCC, FcγRIIIA polymorphism genotyping, measurement of serum cytokine levels, and flow cytometric analysis of immune cell subtypes. Twenty-two patients were enrolled (19 colorectal cancer, 3 HNSCC). Fatigue was the only dose-limiting toxicity. One partial response was observed and 8 patients had stable disease at least 12 weeks. The recommended phase II dose is cetuximab 500 mg/m(2) with lenalidomide 25 mg daily, days 1-21. Correlative studies demonstrated a dose-dependent increase in natural killer cytotoxic activity with increasing doses of lenalidomide. Cetuximab and lenalidomide were well tolerated. There was a lenalidomide dose-dependent increase in ADCC with higher activity in patients enrolled in cohort 3 than those enrolled in cohorts 1/2. Although response was not a primary endpoint, there was evidence of antitumor activity for the combination therapy. Further investigation of lenalidomide as an immunomodulator in solid tumors is warranted. Mol Cancer Ther; 15(9); 2244-50. ©2016 AACR.
Collapse
Affiliation(s)
- Erin M Bertino
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.
| | - Elizabeth L McMichael
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Prashant Trikha
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Melanie Davis
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Bonnie Paul
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Michael Grever
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - William E Carson
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio. Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, Ohio
| | - Gregory A Otterson
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
28
|
McMichael EL, Jaime-Ramirez AC, Guenterberg KD, Luedke E, Atwal LS, Campbell AR, Hu Z, Tatum AS, Kondadasula SV, Mo X, Tridandapani S, Bloomston M, Ellison EC, Williams TM, Bekaii-Saab T, Carson WE. IL-21 Enhances Natural Killer Cell Response to Cetuximab-Coated Pancreatic Tumor Cells. Clin Cancer Res 2016; 23:489-502. [PMID: 27435400 DOI: 10.1158/1078-0432.ccr-16-0004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 06/03/2016] [Accepted: 06/23/2016] [Indexed: 01/29/2023]
Abstract
PURPOSE Alternative strategies to EGFR blockage by mAbs is necessary to improve the efficacy of therapy in patients with locally advanced or metastatic pancreatic cancer. One such strategy includes the use of NK cells to clear cetuximab-coated tumor cells, as need for novel therapeutic approaches to enhance the efficacy of cetuximab is evident. We show that IL-21 enhances NK cell-mediated effector functions against cetuximab-coated pancreatic tumor cells irrespective of KRAS mutation status. EXPERIMENTAL DESIGN NK cells from normal donors or donors with pancreatic cancer were used to assess ADCC, IFN-γ release, and T-cell chemotaxis toward human pancreatic cancer cell lines. The in vivo efficacy of IL-21 in combination with cetuximab was evaluated in a subcutaneous and intraperitoneal model of pancreatic cancer. RESULTS NK cell lysis of cetuximab-coated wild-type and mutant kras pancreatic cancer cell lines were significantly higher following NK cell IL-21 treatment. In response to cetuximab-coated pancreatic tumor cells, IL-21-treated NK cells secreted significantly higher levels of IFN-γ and chemokines, increased chemotaxis of T cells, and enhanced NK cell signal transduction via activation of ERK and STAT1. Treatment of mice bearing subcutaneous or intraperitoneal EGFR-positive pancreatic tumor xenografts with mIL-21 and cetuximab led to significant inhibition of tumor growth, a result further enhanced by the addition of gemcitabine. CONCLUSIONS These results suggest that cetuximab treatment in combination with IL-21 adjuvant therapy in patients with EGFR-positive pancreatic cancer results in significant NK cell activation, irrespective of KRAS mutation status, and may be a potential therapeutic strategy. Clin Cancer Res; 23(2); 489-502. ©2016 AACR.
Collapse
Affiliation(s)
- Elizabeth L McMichael
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio
| | | | - Kristan D Guenterberg
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Eric Luedke
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Lakhvir S Atwal
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Amanda R Campbell
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Zhiwei Hu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Armika S Tatum
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | | | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Susheela Tridandapani
- Division of Pulmonary Medicine, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Mark Bloomston
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio
| | - E Christopher Ellison
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Terence M Williams
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Tanios Bekaii-Saab
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - William E Carson
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio. .,Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
29
|
Collin A, Noacco A, Talvas J, Caldefie-Chézet F, Vasson MP, Farges MC. Enhancement of Lytic Activity by Leptin Is Independent From Lipid Rafts in Murine Primary Splenocytes. J Cell Physiol 2016; 232:101-9. [PMID: 27028718 DOI: 10.1002/jcp.25394] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/25/2016] [Indexed: 12/13/2022]
Abstract
Leptin, a pleiotropic adipokine, is known as a regulator of food intake, but it is also involved in inflammation, immunity, cell proliferation, and survival. Leptin receptor is integrated inside cholesterol-rich microdomains called lipid rafts, which, if disrupted or destroyed, could lead to a perturbation of lytic mechanism. Previous studies also reported that leptin could induce membrane remodeling. In this context, we studied the effect of membrane remodeling in lytic activity modulation induced by leptin. Thus, primary mouse splenocytes were incubated with methyl-β-cyclodextrin (β-MCD), a lipid rafts disrupting agent, cholesterol, a major component of cell membranes, or ursodeoxycholic acid (UDCA), a membrane stabilizer agent for 1 h. These treatments were followed by splenocyte incubation with leptin (absence, 10 and 100 ng/ml). Unlike β-MCD or cholesterol, UDCA was able to block leptin lytic induction. This result suggests that leptin increased the lytic activity of primary spleen cells against syngenic EO771 mammary cancer cells independently from lipid rafts but may involve membrane fluidity. Furthermore, natural killer cells were shown to be involved in the splenocyte lytic activity. To our knowledge it is the first publication in primary culture that provides the link between leptin lytic modulation and membrane remodeling. J. Cell. Physiol. 232: 101-109, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aurore Collin
- Clermont Université, Université d'Auvergne, UFR Pharmacie, UMR 1019, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, Clermont-Ferrand, France. .,INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France.
| | - Audrey Noacco
- Clermont Université, Université d'Auvergne, UFR Pharmacie, UMR 1019, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, Clermont-Ferrand, France.,INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Jérémie Talvas
- Clermont Université, Université d'Auvergne, UFR Pharmacie, UMR 1019, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, Clermont-Ferrand, France.,INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Florence Caldefie-Chézet
- Clermont Université, Université d'Auvergne, UFR Pharmacie, UMR 1019, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, Clermont-Ferrand, France.,INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Marie-Paule Vasson
- Clermont Université, Université d'Auvergne, UFR Pharmacie, UMR 1019, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, Clermont-Ferrand, France.,INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France.,Centre Jean-Perrin, CHU Gabriel-Montpied, Unité de Nutrition, Clermont-Ferrand, France
| | - Marie-Chantal Farges
- Clermont Université, Université d'Auvergne, UFR Pharmacie, UMR 1019, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, Clermont-Ferrand, France.,INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| |
Collapse
|
30
|
Jaime-Ramirez AC, McMichael E, Kondadasula S, Skinner CC, Mundy-Bosse BL, Luedke E, Jones NB, Mani A, Roda J, Karpa V, Li H, Li J, Elavazhagan S, La Perle KM, Schmitt AC, Lu Y, Zhang X, Pan X, Mao H, Davis M, Jarjoura D, Butchar JP, Poi M, Phelps M, Tridandapani S, Byrd JC, Caligiuri MA, Lee RJ, Carson WE. NK Cell-Mediated Antitumor Effects of a Folate-Conjugated Immunoglobulin Are Enhanced by Cytokines. Cancer Immunol Res 2016; 4:323-336. [PMID: 26865456 DOI: 10.1158/2326-6066.cir-15-0168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/28/2015] [Indexed: 12/21/2022]
Abstract
Optimally effective antitumor therapies would not only activate immune effector cells but also engage them at the tumor. Folate conjugated to immunoglobulin (F-IgG) could direct innate immune cells with Fc receptors to folate receptor-expressing cancer cells. F-IgG bound to human KB and HeLa cells, as well as murine L1210JF, a folate receptor (FR)-overexpressing cancer cell line, as determined by flow cytometry. Recognition of F-IgG by natural killer (NK) cell Fc receptors led to phosphorylation of the ERK transcription factor and increased NK cell expression of CD69. Lysis of KB tumor cells by NK cells increased by about 5-fold after treatment with F-IgG, an effect synergistically enhanced by treatment with IL2, IL12, IL15, or IL21 (P< 0.001). F-IgG also enhanced the lysis of chronic lymphocytic leukemia cells by autologous NK cells. NK cells significantly increased production of IFNγ, MIP-1α, and RANTES in response to F-IgG-coated KB target cells in the presence of the NK cell-activating cytokine IL12, and these coculture supernatants induced significant T-cell chemotaxis (P< 0.001). F-IgG-coated targets also stimulated FcR-mediated monocyte effector functions. Studies in a murine leukemia model confirmed the intratumoral localization and antitumor activity of F-IgG, as well as enhancement of its effects by IL12 (P =0.05). The antitumor effect of this combination was dependent on NK cells and led to decreased tumor cell proliferation in vivo Thus, F-IgG can induce an immune response against FR-positive tumor cells that is mediated by NK cells and can be augmented by cytokine therapy.
Collapse
Affiliation(s)
| | - Elizabeth McMichael
- Department of Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH
| | | | | | - Bethany L Mundy-Bosse
- Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Eric Luedke
- Department of Surgery, The Ohio State University, Columbus, OH
| | | | - Aruna Mani
- Breast Cancer Center, Memorial Cancer Institute, Pembroke Pines, FL
| | - Julie Roda
- OncoMed Pharmaceuticals Inc., Redwood City, CA
| | | | - Hong Li
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Jilong Li
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Saranya Elavazhagan
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Krista M La Perle
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH
| | | | | | - Xiaoli Zhang
- Center for Biostatistics, The Ohio State University, Columbus, OH
| | - Xueliang Pan
- Center for Biostatistics, The Ohio State University, Columbus, OH
| | - Hsaioyin Mao
- Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Melanie Davis
- Division of Hematology, The Ohio State University, Columbus, OH
| | - David Jarjoura
- Center for Biostatistics, The Ohio State University, Columbus, OH
| | - Jonathan P Butchar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Ming Poi
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Mitch Phelps
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Susheela Tridandapani
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - John C Byrd
- Division of Hematology, The Ohio State University, Columbus, OH
| | - Michael A Caligiuri
- Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Robert J Lee
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - William E Carson
- Department of Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH.,Department of Surgery, The Ohio State University, Columbus, OH.,Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| |
Collapse
|
31
|
Ren L, Campbell A, Fang H, Gautam S, Elavazhagan S, Fatehchand K, Mehta P, Stiff A, Reader BF, Mo X, Byrd JC, Carson WE, Butchar JP, Tridandapani S. Analysis of the Effects of the Bruton's tyrosine kinase (Btk) Inhibitor Ibrutinib on Monocyte Fcγ Receptor (FcγR) Function. J Biol Chem 2015; 291:3043-52. [PMID: 26627823 DOI: 10.1074/jbc.m115.687251] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Indexed: 02/04/2023] Open
Abstract
The irreversible Bruton's tyrosine kinase (Btk) inhibitor ibrutinib has shown efficacy against B-cell tumors such as chronic lymphocytic leukemia and B-cell non-Hodgkin lymphoma. Fcγ receptors (FcγR) on immune cells such as macrophages play an important role in tumor-specific antibody-mediated immune responses, but many such responses involve Btk. Here we tested the effects of ibrutinib on FcγR-mediated activities in monocytes. We found that ibrutinib did not affect monocyte FcγR-mediated phagocytosis, even at concentrations higher than those achieved physiologically, but suppressed FcγR-mediated cytokine production. We confirmed these findings in macrophages from Xid mice in which Btk signaling is defective. Because calcium flux is a major event downstream of Btk, we tested whether it was involved in phagocytosis. The results showed that blocking intracellular calcium flux decreased FcγR-mediated cytokine production but not phagocytosis. To verify this, we measured activation of the GTPase Rac, which is responsible for actin polymerization. Results showed that ibrutinib did not inhibit Rac activation, nor did the calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester). We next asked whether the effect of ibrutinib on monocyte FcγR-mediated cytokine production could be rescued by IFNγ priming because NK cells produce IFNγ in response to antibody therapy. Pretreatment of monocytes with IFNγ abrogated the effects of ibrutinib on FcγR-mediated cytokine production, suggesting that IFNγ priming could overcome this Btk inhibition. Furthermore, in monocyte-natural killer cell co-cultures, ibrutinib did not inhibit FcγR-mediated cytokine production despite doing so in single cultures. These results suggest that combining ibrutinib with monoclonal antibody therapy could enhance chronic lymphocytic leukemia cell killing without affecting macrophage effector function.
Collapse
Affiliation(s)
- Li Ren
- From the Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun 130000, China and
| | | | | | | | | | | | | | | | | | - Xiaokui Mo
- Center for Biostatistics, Ohio State University, Columbus, Ohio 43210
| | | | | | | | | |
Collapse
|
32
|
Campbell AR, Regan K, Bhave N, Pattanayak A, Parihar R, Stiff AR, Trikha P, Scoville SD, Liyanarachchi S, Kondadasula SV, Lele O, Davuluri R, Payne PRO, Carson WE. Gene expression profiling of the human natural killer cell response to Fc receptor activation: unique enhancement in the presence of interleukin-12. BMC Med Genomics 2015; 8:66. [PMID: 26470881 PMCID: PMC4608307 DOI: 10.1186/s12920-015-0142-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 10/07/2015] [Indexed: 01/23/2023] Open
Abstract
Background Traditionally, the CD56dimCD16+ subset of Natural Killer (NK) cells has been thought to mediate cellular cytotoxicity with modest cytokine secretion capacity. However, studies have suggested that this subset may exert a more diverse array of immunological functions. There exists a lack of well-developed functional models to describe the behavior of activated NK cells, and the interactions between signaling pathways that facilitate effector functions are not well understood. In the present study, a combination of genome-wide microarray analyses and systems-level bioinformatics approaches were utilized to elucidate the transcriptional landscape of NK cells activated via interactions with antibody-coated targets in the presence of interleukin-12 (IL-12). Methods We conducted differential gene expression analysis of CD56dimCD16+ NK cells following FcR stimulation in the presence or absence of IL-12. Next, we functionally characterized gene sets according to patterns of gene expression and validated representative genes using RT-PCR. IPA was utilized for biological pathway analysis, and an enriched network of interacting genes was generated using GeneMANIA. Furthermore, PAJEK and the HITS algorithm were employed to identify important genes in the network according to betweeness centrality, hub, and authority node metrics. Results Analyses revealed that CD56dimCD16+ NK cells co-stimulated via the Fc receptor (FcR) and IL-12R led to the expression of a unique set of genes, including genes encoding cytotoxicity receptors, apoptotic proteins, intracellular signaling molecules, and cytokines that may mediate enhanced cytotoxicity and interactions with other immune cells within inflammatory tissues. Network analyses identified a novel set of connected key players, BATF, IRF4, TBX21, and IFNG, within an integrated network composed of differentially expressed genes in NK cells stimulated by various conditions (immobilized IgG, IL-12, or the combination of IgG and IL-12). Conclusions These results are the first to address the global mechanisms by which NK cells mediate their biological functions when encountering antibody-coated targets within inflammatory sites. Moreover, this study has identified a set of high-priority targets for subsequent investigation into strategies to combat cancer by enhancing the anti-tumor activity of CD56dimCD16+ NK cells. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0142-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amanda R Campbell
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA. .,Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, 43210, USA.
| | - Kelly Regan
- Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, 43210, USA. .,Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA.
| | - Neela Bhave
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA.
| | - Arka Pattanayak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA.
| | - Robin Parihar
- Department of Pediatrics, The Cleveland Clinic, Cleveland, OH, 44106, USA.
| | - Andrew R Stiff
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA. .,Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, 43210, USA.
| | - Prashant Trikha
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA.
| | - Steven D Scoville
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA. .,Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, 43210, USA.
| | - Sandya Liyanarachchi
- Division of Human Cancer Genetics, The Ohio State University, Columbus, OH, 43210, USA.
| | - Sri Vidya Kondadasula
- Departments of Oncology and Medicine, Wayne State University and Barbara Ann Karmanos Cancer Institute, Detroit, MI, 48201, USA.
| | - Omkar Lele
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA.
| | - Ramana Davuluri
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA.
| | - Philip R O Payne
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA.
| | - William E Carson
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA. .,Department of Surgery, The Ohio State University, Columbus, OH, 43210, USA. .,The Ohio State University College of Medicine, N924 Doan Hall, 410 West 10th Ave., Columbus, OH, 43210, USA.
| |
Collapse
|
33
|
Liu ZG, Jiao XY, Chen ZG, Feng K, Luo HH. Estrogen receptorβ2 regulates interlukin-12 receptorβ2 expression via p38 mitogen-activated protein kinase signaling and inhibits non-small-cell lung cancer proliferation and invasion. Mol Med Rep 2015; 12:248-54. [PMID: 25695486 DOI: 10.3892/mmr.2015.3366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 07/25/2014] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is one of the most common types of cancer and is the leading cause of cancer-related mortality worldwide. Estrogens are known to be involved in the development and progression of non-small-cell lung cancer (NSCLC). These effects are initially mediated through binding of estrogen to estrogen receptors (ERs), in particular ERβ2. Our preliminary studies demonstrated that ERβ2 and interleukin-12 receptorβ2 (IL-12Rβ2) expression are correlated in NSCLC. The present study investigated the expression of these proteins in NSCLC cells and how changes in their expression affected cell proliferation and invasion. In addition, it aimed to explore whether p38 mitogen-activated protein kinase (p38MAPK) is involved in the regulation of IL-12Rβ2 expression by ERβ2. An immunocytochemical array was used to observe the distribution of ERβ2 and IL-12Rβ2. Co-immuoprecipitation was employed to observe the interaction between p38MAPK and IL-12Rβ2, by varying the expression of ERβ2 and p38MAPK. Western-blot analysis and reverse transcription-polymerase chain reaction assays were used to investigate the mechanism underlying ERβ2 regulation of IL-12Rβ2 expression. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, scratch wound healing and Transwell assays were used to investigate the impact of ERβ2 on proliferative, invasive and migratory abilities of NSCLC cells. ERβ2 was predominantly found in the cytoplasm and nucleus, whilst IL-12Rβ2 was largely confined to the cytoplasm, although a degree of expression was observed in the nucleus. Compared with normal bronchial epithelial cells, IL-12Rβ2 and ERβ2 were overexpressed in the NSCLC cell groups. Coimmuoprecipitation demonstrated an interaction between p38MAPK and IL-12Rβ2. ERβ2 appeared to upregulate IL-12Rβ2 expression and inhibition of p38MAPK attenuated this effect. ERβ2 and IL-12Rβ2 expression inhibited the proliferation, metastasis and invasion of NSCLC cell lines, but knockout of IL-12Rβ2, even in the presence of ERβ2, led to an increase in NSCLC cell proliferation and invasiveness. In conclusion, to the best of our knowledge this study is the first to demonstrate that IL-12Rβ2 may be important in the mechanisms underlying ERβ2 inhibition of NSCLC development, and that this interaction may be mediated via p38MAPK.
Collapse
Affiliation(s)
- Zhao-Guo Liu
- Department of General Thoracic Surgery, First Affiliated Hospital, Sun‑Yat sen University, Guangzhou, Guangdong 510089, P.R. China
| | - Xing-Yuan Jiao
- Organ Transplantation Center, First Affiliated Hospital, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhen-Guang Chen
- Department of General Thoracic Surgery, First Affiliated Hospital, Sun‑Yat sen University, Guangzhou, Guangdong 510089, P.R. China
| | - Ke Feng
- Department of General Thoracic Surgery, First Affiliated Hospital, Sun‑Yat sen University, Guangzhou, Guangdong 510089, P.R. China
| | - Hong-He Luo
- Department of General Thoracic Surgery, First Affiliated Hospital, Sun‑Yat sen University, Guangzhou, Guangdong 510089, P.R. China
| |
Collapse
|
34
|
Balance of cellular and humoral immunity determines the level of protection by HIV vaccines in rhesus macaque models of HIV infection. Proc Natl Acad Sci U S A 2015; 112:E992-9. [PMID: 25681373 DOI: 10.1073/pnas.1423669112] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A guiding principle for HIV vaccine design has been that cellular and humoral immunity work together to provide the strongest degree of efficacy. However, three efficacy trials of Ad5-vectored HIV vaccines showed no protection. Transmission was increased in two of the trials, suggesting that this vaccine strategy elicited CD4+ T-cell responses that provide more targets for infection, attenuating protection or increasing transmission. The degree to which this problem extends to other HIV vaccine candidates is not known. Here, we show that a gp120-CD4 chimeric subunit protein vaccine (full-length single chain) elicits heterologous protection against simian-human immunodeficiency virus (SHIV) or simian immunodeficiency virus (SIV) acquisition in three independent rhesus macaque repeated low-dose rectal challenge studies with SHIV162P3 or SIVmac251. Protection against acquisition was observed with multiple formulations and challenges. In each study, protection correlated with antibody-dependent cellular cytotoxicity specific for CD4-induced epitopes, provided that the concurrent antivaccine T-cell responses were minimal. Protection was lost in instances when T-cell responses were high or when the requisite antibody titers had declined. Our studies suggest that balance between a protective antibody response and antigen-specific T-cell activation is the critical element to vaccine-mediated protection against HIV. Achieving and sustaining such a balance, while enhancing antibody durability, is the major challenge for HIV vaccine development, regardless of the immunogen or vaccine formulation.
Collapse
|
35
|
Gütgemann SA, Sandusky MM, Wingert S, Claus M, Watzl C. Recruitment of activating NK-cell receptors 2B4 and NKG2D to membrane microdomains in mammalian cells is dependent on their transmembrane regions. Eur J Immunol 2015; 45:1258-69. [DOI: 10.1002/eji.201444741] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 11/21/2014] [Accepted: 12/22/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Stephan A. Gütgemann
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| | - Mina M. Sandusky
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| | - Sabine Wingert
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| | - Maren Claus
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| | - Carsten Watzl
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| |
Collapse
|
36
|
Ziblat A, Domaica CI, Spallanzani RG, Iraolagoitia XLR, Rossi LE, Avila DE, Torres NI, Fuertes MB, Zwirner NW. IL-27 stimulates human NK-cell effector functions and primes NK cells for IL-18 responsiveness. Eur J Immunol 2014; 45:192-202. [PMID: 25308526 DOI: 10.1002/eji.201444699] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 09/10/2014] [Accepted: 10/06/2014] [Indexed: 12/27/2022]
Abstract
IL-27, a member of the IL-12 family of cytokines, is produced by APCs, and displays pro- and anti-inflammatory effects. How IL-27 affects human NK cells still remains unknown. In this study, we observed that mature DCs secreted IL-27 and that blockade of IL-27R (CD130) reduced the amount of IFN-γ produced by NK cells during their coculture, showing the importance of IL-27 during DC-NK-cell crosstalk. Accordingly, human rIL-27 stimulated IFN-γ secretion by NK cells in a STAT1-dependent manner, induced upregulation of CD25 and CD69 on NK cells, and displayed a synergistic effect with IL-18. Preincubation experiments demonstrated that IL-27 primed NK cells for IL-18-induced IFN-γ secretion, which was associated with an IL-27-driven upregulation of T-bet expression. Also, IL-27 triggered NKp46-dependent NK-cell-mediated cytotoxicity against Raji, T-47D, and HCT116 cells, and IL-18 enhanced this cytotoxic response. Such NK-cell-mediated cytotoxicity involved upregulation of perforin, granule exocytosis, and TRAIL-mediated cytotoxicity but not Fas-FasL interaction. Moreover, IL-27 also potentiated Ab-dependent cell-mediated cytotoxicity against mAb-coated target cells. Taken together, IL-27 stimulates NK-cell effector functions, which might be relevant in different physiological and pathological situations.
Collapse
Affiliation(s)
- Andrea Ziblat
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Köther K, Nordhoff C, Masemann D, Varga G, Bream JH, Gaestel M, Wixler V, Ludwig S. MAPKAP kinase 3 suppresses Ifng gene expression and attenuates NK cell cytotoxicity and Th1 CD4 T-cell development upon influenza A virus infection. FASEB J 2014; 28:4235-46. [PMID: 24935968 DOI: 10.1096/fj.14-249599] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
MK2 and MK3 are downstream targets of p38 and ERK1/2. They control the mRNA stability of several inflammatory cytokines, including TNF-α and IL-10. Whereas MK2 is expressed ubiquitously, the expression of MK3 is restricted to muscle, liver, and heart tissues and T and NK cells. Using Mk-deficient and wild-type (WT) mice, we demonstrated an inhibitory effect of MK3, but not of MK2, on interferon (IFN)-γ expression in T and NK lymphocytes. The results provided evidence that the inhibitory effect of MK3 is based on negative feedback phosphorylation of p38 and ERK1/2, which causes decreased binding of Stat4 to the IFN-γ promoter and reduced expression of IFN-γ mRNA and protein. Consequently, all Mk3(-/-) mice challenged with the Th1-inducing influenza A virus (IAV) survived the WT LD50 virus dose. The reduced disease severity in the Mk3(-/-) mice was accompanied by a >10-fold reduction in viral lung titer and an increase in the number of activated NK cells and enhanced Th1 activation of CD4 T cells. Thus, our data describe the protein kinase MK3 as a novel regulator of the innate and adaptive immune responses.-Köther, K., Nordhoff, C., Masemann, D., Varga, G., Bream, J. H., Gaestel, M., Wixler, V., Ludwig, S. MAPKAP kinase 3 suppresses Ifng gene expression and attenuates NK cell cytotoxicity and Th1 CD4 T-cell development upon influenza A virus infection.
Collapse
Affiliation(s)
- Katharina Köther
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), and
| | - Carolin Nordhoff
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), and
| | - Dörthe Masemann
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), and
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, Westfälische Wilhelms University, Münster, Germany
| | - Jay H Bream
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Matthias Gaestel
- Institute of Biochemistry, Hannover Medical School, Hannover, Germany; and
| | - Viktor Wixler
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), and Interdisciplinary Center of Clinical Research (IZKF), University of Münster, Münster, Germany
| | - Stephan Ludwig
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), and Interdisciplinary Center of Clinical Research (IZKF), University of Münster, Münster, Germany
| |
Collapse
|
38
|
Serrano-Pertierra E, Cernuda-Morollón E, Brdička T, Hoøejši V, López-Larrea C. L-plastin is involved in NKG2D recruitment into lipid rafts and NKG2D-mediated NK cell migration. J Leukoc Biol 2014; 96:437-45. [PMID: 24803550 DOI: 10.1189/jlb.2a1013-564r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Membrane rafts are microdomains of the plasma membrane that have multiple biological functions. The involvement of these structures in the biology of T cells, namely in signal transduction by the TCR, has been widely studied. However, the role of membrane rafts in immunoreceptor signaling in NK cells is less well known. We studied the distribution of the activating NKG2D receptor in lipid rafts by isolating DRMs in a sucrose density gradient or by raft fractionation by β-OG-selective solubility in the NKL cell line. We found that the NKG2D-DAP10 complex and pVav are recruited into rafts upon receptor stimulation. Qualitative proteomic analysis of these fractions showed that the actin cytoskeleton is involved in this process. In particular, we found that the actin-bundling protein L-plastin plays an important role in the clustering of NKG2D into lipid rafts. Moreover, coengagement of the inhibitory receptor NKG2A partially disrupted NKG2D recruitment into rafts. Furthermore, we demonstrated that L-plastin participates in NKG2D-mediated inhibition of NK cell chemotaxis.
Collapse
Affiliation(s)
| | - Eva Cernuda-Morollón
- Neurology Departments, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Tomáš Brdička
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic; and
| | - Václav Hoøejši
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic; and
| | | |
Collapse
|
39
|
Bauman JE, Ferris RL. Integrating novel therapeutic monoclonal antibodies into the management of head and neck cancer. Cancer 2014; 120:624-32. [PMID: 24222079 PMCID: PMC4095869 DOI: 10.1002/cncr.28380] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/21/2013] [Accepted: 08/23/2013] [Indexed: 12/12/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an immunosuppressive malignancy. Interest in developing novel immunotherapies in HNSCC has been reawakened by the success of cetuximab, a therapeutic monoclonal antibody (mAb) against the epidermal growth factor receptor, which likely relies on immune as well as antisignaling mechanisms. This review focuses on novel therapeutic mAbs in current clinical development against established mechanisms of immune evasion in HNSCC, targeting: 1) tumor antigens, with resultant potential to induce antibody-dependent cell-mediated cytotoxicity and T cell activation; 2) immunosuppressive cytokines; 3) costimulatory tumor necrosis factor-family receptors; and 4) coinhibitory immune checkpoint receptors. Clinical trials of immunotherapeutic mAbs as monotherapy, in combination with cytolytic standard therapies exposing tumor antigens or in combination with other immunomodulatory mAbs, are urgently needed in HNSCC.
Collapse
MESH Headings
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antibody-Dependent Cell Cytotoxicity/immunology
- Antigens, Neoplasm/drug effects
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/immunology
- Cetuximab
- Cytokines/antagonists & inhibitors
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/immunology
- Humans
- Immunoglobulin G/drug effects
- Immunotherapy/methods
- Receptors, Tumor Necrosis Factor/antagonists & inhibitors
- Squamous Cell Carcinoma of Head and Neck
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Julie E Bauman
- Department of Internal Medicine (Division of Hematology/Oncology), University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | | |
Collapse
|
40
|
Romee R, Leong JW, Fehniger TA. Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer. SCIENTIFICA 2014; 2014:205796. [PMID: 25054077 PMCID: PMC4099226 DOI: 10.1155/2014/205796] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 05/02/2014] [Indexed: 05/11/2023]
Abstract
Natural killer (NK) cells are innate lymphoid cells important for host defense against pathogens and mediate antitumor immunity. Cytokine receptors transduce important signals that regulate proliferation, survival, activation status, and trigger effector functions. Here, we review the roles of major cytokines that regulate human NK cell development, survival, and function, including IL-2, IL-12, IL-15, IL-18, and IL-21, and their translation to the clinic as immunotherapy agents. We highlight a recent development in NK cell biology, the identification of innate NK cell memory, and focus on cytokine-induced memory-like (CIML) NK cells that result from a brief, combined activation with IL-12, IL-15, and IL-18. This activation results in long lived NK cells that exhibit enhanced functionality when they encounter a secondary stimulation and provides a new approach to enable NK cells for enhanced responsiveness to infection and cancer. An improved understanding of the cellular and molecular aspects of cytokine-cytokine receptor signals has led to a resurgence of interest in the clinical use of cytokines that sustain and/or activate NK cell antitumor potential. In the future, such strategies will be combined with negative regulatory signal blockade and enhanced recognition to comprehensively enhance NK cells for immunotherapy.
Collapse
Affiliation(s)
- Rizwan Romee
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey W. Leong
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Todd A. Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- *Todd A. Fehniger:
| |
Collapse
|
41
|
Gildener-Leapman N, Ferris RL, Bauman JE. Promising systemic immunotherapies in head and neck squamous cell carcinoma. Oral Oncol 2013; 49:1089-96. [PMID: 24126223 PMCID: PMC4861147 DOI: 10.1016/j.oraloncology.2013.09.009] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 01/11/2023]
Abstract
Patients with head and neck squamous cell carcinoma (HNSCC) demonstrate poor survival and significant treatment morbidity with standard therapy. The immune profile in HNSCC, whether caused by carcinogen exposure or human papillomavirus (HPV), is notably immunosuppressive. Early clinical trials of immunotherapy in HNSCC were troubled by systemic toxicity or difficulties in local administration. Now, interest in immunotherapy has been revitalized by mechanistic insights into immune evasion by HNSCC, coupled to ongoing development of novel immunotherapies. This review will summarize immune escape mechanisms in HNSCC, namely downregulation of tumor antigen (TA) presentation, aberrant regulation of the signal transducer and activator of transcription (STAT) family, the immunosuppressive cytokine milieu, and dysregulation of immune effector cells. Therapeutic strategies hypothesized to specifically counter HNSCC immunosuppression will then be discussed. We will survey TA- targeted monoclonal antibodies (mAb), including the prototype cetuximab, as well as adjunctive strategies to enhance antibody-dependent cell-mediated cytotoxicity. We will review immunomodulation to restore STAT1/STAT3 activation balance. Examples of mAb therapy to block immunosuppressive cytokines, such as interleukin-6 or VEGF, will be provided. mAbs which release co-inhibitory T cell receptors such as CTLA-4 and PD-1, overexpressed in HNSCC, also hold therapeutic promise. Finally, we will describe principles for therapeutic vaccination in HPV-associated HNSCC, where non-host TAs such as viral oncoproteins represent ideal targets, and HPV-negative HNSCC, where p53 is a promising target. Insights into immunosuppression in HNSCC have elucidated mechanistic targets for immunotherapy. Rational clinical investigation may lead to effective stand alone or combinatorial treatment approaches.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Antigens, Neoplasm/drug effects
- Antigens, Neoplasm/immunology
- B-Lymphocyte Subsets/drug effects
- B-Lymphocyte Subsets/immunology
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/therapy
- Cytokines/drug effects
- Cytokines/immunology
- Genes, p53/drug effects
- Genes, p53/immunology
- Head and Neck Neoplasms/immunology
- Head and Neck Neoplasms/therapy
- Humans
- Immunotherapy/methods
- Papillomaviridae/drug effects
- Papillomaviridae/immunology
- Receptors, Antigen, T-Cell/drug effects
- Receptors, Antigen, T-Cell/immunology
- STAT Transcription Factors/drug effects
- STAT Transcription Factors/immunology
Collapse
Affiliation(s)
- Neil Gildener-Leapman
- Department of Otolaryngology, University of Pittsburgh Eye and Ear Institute, 203 Lothrop Street, Pittsburgh, PA 15213, United States
| | - Robert L. Ferris
- Department of Otolaryngology, University of Pittsburgh Eye and Ear Institute, 203 Lothrop Street, Pittsburgh, PA 15213, United States
| | - Julie E. Bauman
- Department of Internal Medicine, University of Pittsburgh Cancer Institute, 5115 Centre Avenue 2nd Floor, Pittsburgh, PA 15232, United States
| |
Collapse
|
42
|
Padro CJ, Shawler TM, Gormley MG, Sanders VM. Adrenergic regulation of IgE involves modulation of CD23 and ADAM10 expression on exosomes. THE JOURNAL OF IMMUNOLOGY 2013; 191:5383-97. [PMID: 24140643 DOI: 10.4049/jimmunol.1301019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Soluble CD23 plays a role in the positive regulation of an IgE response. Engagement of the β2 adrenergic receptor (β2AR) on a B cell is known to enhance the level of both soluble CD23 and IgE, although the mechanism by which this occurs is not completely understood. In this study, we report that, in comparison with a CD40 ligand/IL-4-primed murine B cell alone, β2AR engagement on a primed B cell increased gene expression of a disintegrin and metalloproteinase (ADAM)10, which is the primary sheddase of CD23, as well as protein expression of both CD23 and ADAM10, in a protein kinase A- and p38 MAPK-dependent manner, and promoted the localization of these proteins to exosomes as early as 2 d after priming, as determined by both Western blot and flow cytometry and confirmed by electron microscopy. In comparison with isolated exosomes released from primed B cells alone, the transfer of exosomes released from β2AR agonist-exposed primed B cells to cultures of recipient primed B cells resulted in an increase in the level of IgE produced per cell, without affecting the number of cells producing IgE, as determined by ELISPOT. These effects still occurred when a β2AR antagonist was added along with the transfer to block residual agonist, and they failed to occur when exosomes were isolated from β2AR-deficient B cells. These findings suggest that the mechanism responsible for mediating the β2AR-induced increase in IgE involves a shuttling of the β2AR-induced increase in CD23 and ADAM10 proteins to exosomes that subsequently mediate an increase in IgE.
Collapse
Affiliation(s)
- Caroline J Padro
- Biomedical Sciences Graduate Program, The Ohio State University Wexner College of Medicine, Columbus, OH 43210
| | | | | | | |
Collapse
|
43
|
Srivastava S, Pelloso D, Feng H, Voiles L, Lewis D, Haskova Z, Whitacre M, Trulli S, Chen YJ, Toso J, Jonak ZL, Chang HC, Robertson MJ. Effects of interleukin-18 on natural killer cells: costimulation of activation through Fc receptors for immunoglobulin. Cancer Immunol Immunother 2013; 62:1073-82. [PMID: 23604103 PMCID: PMC3707624 DOI: 10.1007/s00262-013-1403-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 02/08/2013] [Indexed: 11/30/2022]
Abstract
The antitumor activity of monoclonal antibodies is mediated by effector cells, such as natural killer (NK) cells, that express Fc receptors for immunoglobulin. Efficacy of monoclonal antibodies, including the CD20 antibody rituximab, could be improved by agents that augment the function of NK cells. Interleukin (IL)-18 is an immunostimulatory cytokine that has antitumor activity in preclinical models. The effects of IL-18 on NK cell function mediated through Fcγ receptors were examined. Human NK cells stimulated with immobilized IgG in vitro secreted IFN-γ as expected; such IFN-γ production was partially inhibited by blocking CD16 with monoclonal antibodies. IL-18 augmented IFN-γ production by NK cells stimulated with immobilized IgG or CD16 antibodies. NK cell IFN-γ production in response to immobilized IgG and/or IL-18 was inhibited by chemical inhibitors of Syk and several other kinases involved in CD16 signaling pathways. IL-18 augmented antibody-dependent cellular cytotoxicity (ADCC) of human NK cells against rituximab-coated Raji cells in vitro. IL-18 and rituximab acted synergistically to promote regression of human lymphoma xenografts in SCID mice. Inasmuch as IL-18 costimulates IFN-γ production and ADCC of NK cells activated through Fc receptors in vitro and augments antitumor activity of rituximab in vivo, it is an attractive cytokine to combine with monoclonal antibodies for treatment of human cancer.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal, Murine-Derived/administration & dosage
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antibody-Dependent Cell Cytotoxicity/immunology
- Antineoplastic Agents/administration & dosage
- Cell Line, Tumor
- Disease Models, Animal
- Female
- Humans
- Immunoglobulin G/immunology
- Immunoglobulins/metabolism
- Interferon-gamma/biosynthesis
- Interleukin-18/administration & dosage
- Interleukin-18/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphoma/drug therapy
- Lymphoma/immunology
- Mice
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/metabolism
- Receptors, Fc/metabolism
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Rituximab
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Shivani Srivastava
- Bone Marrow and Stem Cell Transplantation Program, Indianapolis, IN, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Monoclonal antibody therapy of pancreatic cancer with cetuximab: potential for immune modulation. J Immunother 2013; 35:367-73. [PMID: 22576341 DOI: 10.1097/cji.0b013e3182562d76] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Pancreatic cancer is a devastating disease, with a median survival of around 6 months for patients with stage IV disease. The epidermal growth factor receptor (EGFR, or HER1) belongs to the erbB receptor tyrosine kinase family. HER1-mediated cell signaling has been shown to play a major role in promoting tumor proliferation, angiogenesis, metastasis, and evasion of apoptosis. Over-expression of HER1 is observed in multiple human malignancies, including colorectal, lung, breast and pancreatic cancers. In pancreatic carcinoma, over-expression of HER1 is observed in greater than 70% of patients and is associated with a poor prognosis and a significant decrease in survival. Cetuximab (Erbitux) is a chimeric monoclonal antibody (mAb) that binds to the extracellular domain of the HER1 molecule preventing ligand binding and promoting internalization and subsequent degradation of the HER1 receptor. Cetuximab has shown anti-tumor activity either alone or in combination with other agents and is currently FDA approved for use in both squamous cell carcinoma of the head and neck (SCCHN) and colorectal carcinoma. Research efforts continue to elucidate a possible role for cetuximab in the treatment of pancreatic cancer. Despite promising preclinical work, phase II and phase III clinical trials have failed to consistently show efficacy of cetuximab treatment in advanced pancreatic cancer either alone or in combination with cytotoxic agents. Alternative approaches to HER1 blockade and mAbs including immune modulation with cytokines might be necessary in order to improve the efficacy of mAbs in pancreatic cancer therapy.
Collapse
|
45
|
Mueller M, Barros P, Witherden A, Roberts A, Zhang Z, Schaschl H, Yu CY, Hurles M, Schaffner C, Floto R, Game L, Steinberg K, Wilson R, Graves T, Eichler E, Cook H, Vyse T, Aitman T. Genomic pathology of SLE-associated copy-number variation at the FCGR2C/FCGR3B/FCGR2B locus. Am J Hum Genet 2013; 92:28-40. [PMID: 23261299 PMCID: PMC3542466 DOI: 10.1016/j.ajhg.2012.11.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 09/12/2012] [Accepted: 11/26/2012] [Indexed: 01/18/2023] Open
Abstract
Reduced FCGR3B copy number is associated with increased risk of systemic lupus erythematosus (SLE). The five FCGR2/FCGR3 genes are arranged across two highly paralogous genomic segments on chromosome 1q23. Previous studies have suggested mechanisms for structural rearrangements at the FCGR2/FCGR3 locus and have proposed mechanisms whereby altered FCGR3B copy number predisposes to autoimmunity, but the high degree of sequence similarity between paralogous segments has prevented precise definition of the molecular events and their functional consequences. To pursue the genomic pathology associated with FCGR3B copy-number variation, we integrated sequencing data from fosmid and bacterial artificial chromosome clones and sequence-captured DNA from FCGR3B-deleted genomes to establish a detailed map of allelic and paralogous sequence variation across the FCGR2/FCGR3 locus. This analysis identified two highly paralogous 24.5 kb blocks within the FCGR2C/FCGR3B/FCGR2B locus that are devoid of nonpolymorphic paralogous sequence variations and that define the limits of the genomic regions in which nonallelic homologous recombination leads to FCGR2C/FCGR3B copy-number variation. Further, the data showed evidence of swapping of haplotype blocks between these highly paralogous blocks that most likely arose from sequential ancestral recombination events across the region. Functionally, we found by flow cytometry, immunoblotting and cDNA sequencing that individuals with FCGR3B-deleted alleles show ectopic presence of FcγRIIb on natural killer (NK) cells. We conclude that FCGR3B deletion juxtaposes the 5'-regulatory sequences of FCGR2C with the coding sequence of FCGR2B, creating a chimeric gene that results in an ectopic accumulation of FcγRIIb on NK cells and provides an explanation for SLE risk associated with reduced FCGR3B gene copy number.
Collapse
Affiliation(s)
- Michael Mueller
- Physiological Genomics and Medicine Group, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Paula Barros
- Department of Medical and Molecular Genetics, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Abigail S. Witherden
- Department of Medical and Molecular Genetics, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Amy L. Roberts
- Department of Medical and Molecular Genetics, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Zhou Zhang
- Physiological Genomics and Medicine Group, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Helmut Schaschl
- Department of Medical and Molecular Genetics, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Chack-Yung Yu
- Center for Molecular and Human Genetics, Nationwide Children’s Hospital and Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | - Matthew E. Hurles
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SA, UK
| | - Catherine Schaffner
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - R. Andres Floto
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Laurence Game
- Genomics Core Laboratory, MRC Clinical Sciences Centre, London W12 0NN, UK
| | - Karyn Meltz Steinberg
- Department of Genome Sciences, University of Washington School of Medicine and the Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Richard K. Wilson
- The Genome Institute at Washington University, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tina A. Graves
- The Genome Institute at Washington University, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Evan E. Eichler
- Department of Genome Sciences, University of Washington School of Medicine and the Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - H. Terence Cook
- Centre for Complement and Inflammation Research, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Timothy J. Vyse
- Department of Medical and Molecular Genetics, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Timothy J. Aitman
- Physiological Genomics and Medicine Group, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| |
Collapse
|
46
|
Control of early Theiler's murine encephalomyelitis virus replication in macrophages by interleukin-6 occurs in conjunction with STAT1 activation and nitric oxide production. J Virol 2012; 86:10841-51. [PMID: 22837198 DOI: 10.1128/jvi.01402-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
During Theiler's murine encephalomyelitis virus (TMEV) infection of macrophages, it is thought that high interleukin-6 (IL-6) levels contribute to the demyelinating disease found in chronically infected SJL/J mice but absent in B10.S mice capable of clearing the infection. Therefore, IL-6 expression was measured in TMEV-susceptible SJL/J and TMEV-resistant B10.S macrophages during their infection with TMEV DA strain or responses to lipopolysaccharide (LPS) or poly(I · C). Unexpectedly, IL-6 production was greater in B10.S macrophages than SJL/J macrophages during the first 24 h after stimulation with TMEV, LPS, or poly(I · C). Further experiments showed that in B10.S, SJL/J, and RAW264.7 macrophage cells, IL-6 expression was dependent on extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) and enhanced by exogenous IL-12. In SJL/J and RAW264.7 macrophages, exogenous IL-6 resulted in decreased TMEV replication, earlier activation of STAT1 and STAT3, production of nitric oxide, and earlier upregulation of several antiviral genes downstream of STAT1. However, neither inhibition of IL-6-induced nitric oxide nor knockdown of STAT1 diminished the early antiviral effect of exogenous IL-6. In addition, neutralization of endogenous IL-6 from SJL/J macrophages with Fab antibodies did not exacerbate early TMEV infection. Therefore, endogenous IL-6 expression after TMEV infection is dependent on ERK MAPK, enhanced by IL-12, but too slow to decrease viral replication during early infection. In contrast, exogenous IL-6 enhances macrophage control of TMEV infection through preemptive antiviral nitric oxide production and antiviral STAT1 activation. These results indicate that immediate-early production of IL-6 could protect macrophages from TMEV infection.
Collapse
|
47
|
Luedke E, Jaime-Ramirez AC, Bhave N, Roda J, Choudhary MM, Kumar B, Teknos TN, Carson WE. Cetuximab therapy in head and neck cancer: immune modulation with interleukin-12 and other natural killer cell-activating cytokines. Surgery 2012; 152:431-40. [PMID: 22770960 DOI: 10.1016/j.surg.2012.05.035] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 05/14/2012] [Indexed: 11/16/2022]
Abstract
BACKGROUND Squamous cell carcinoma of the head and neck (SCCHN) is the sixth most common cancer worldwide. Greater than 90% of SCCHN of the oropharynx overexpress the epidermal growth factor receptor (EGFR or HER1). Cetuximab (Erbitux-TM) is a humanized anti-HER1 monoclonal antibody (mAb) that binds to HER1 overexpressing tumor cells. Cetuximab has a direct effect on HER1-positive cancer cells, but it also can activate immune cells that bear receptors for the Fc (constant portion) of IgG such as natural killer (NK) cells. NK cells have an activating Fc receptor for IgG (FcγRIIIa), which mediates Ab dependent cellular cytotoxicity (ADCC) and enhances production of interferon-γ (IFN-γ) in response to Ab-coated targets. Interleukin-12 (IL-12) is a cytokine produced by antigen-presenting cells that stimulates IFN-γ production from NK cells. We hypothesized that IL-12 would enhance the anti-tumor activity of cetuximab by activating the FcR effector mechanisms of NK cells. METHODS Expression of HER1 was measured on human papilloma virus (HPV)-positive (UD-SCC2, UM-SCC47) and HPV-negative (Cal27, UM-SCC74B) SCCHN cell lines by immunoblot analysis and flow cytometry. NK cells from normal donors were treated overnight with IL-2 (100 U), IL-12, IL-15, or IL-21 (all 10 ng/mL) and tested for ADCC versus cetuximab-coated cancer cells in a 4 hr (51)Cr assay. Release of cytokines by NK cells in response to cetuximab-coated cells was measured by ELISA. Phosphorylation of the ERK transcription factor in NK cells was measured by flow cytometry. The efficacy of combination therapy with cetuximab plus IL-12 was evaluated in a murine tumor model of head and neck cancer. RESULTS All cell lines showed >99% expression of HER1 by flow cytometry and immunoblot analysis except UM-SCC74B (73%). Normal NK cells mediated 49.4% lysis of cetuximab-coated SCCHN cell lines as compared to 7.6% lysis of cells treated with control IgG (P = .0002). NK cell lysis of cetuximab-coated SCCHN cells was markedly enhanced by 12 hr pre-treatment of NK cells with IL-12 (71.6% lysis, P = .005 vs cetuximab alone). As a control, IL-12-activated NK cells were tested against IgG-treated cells. ADCC under these conditions was just 21.7%. Similar levels of lysis were noted for both HPV-positive and HPV-negative and cell lines. Other NK cell activating factors such as IL-2, IL-15, and IL-21 were also able to enhance NK cell ADCC. The stimulus of IL-12 and cetuximab-coated tumor cells induced the synergistic production of nanogram levels of IFN-γ (>6-fold increase over controls) (P < .001). A similar effect was seen for NK cell production of the chemokines RANTES, MIP-1α, and IL-8. Phosphorylation of ERK (which is critical for FcR-mediated ADCC and cytokine production) was enhanced in NK cells exposed to IL-12 and IgG as compared to control conditions. The combination of cetuximab plus IL-12 resulted in a reduction in tumor burden when compared to either agent alone in a murine xenograft model of SCCHN. CONCLUSION Cytokine stimulation of NK cells in the presence of cetuximab-coated head and neck cancer cells leads to enhanced NK cell mediated ADCC and cytokine secretion independent of tumor cell HPV-status. Cytokine administration could be a useful adjuvant in the cetuximab treatment of HER1-positive head and neck cancer.
Collapse
Affiliation(s)
- Eric Luedke
- Department of Surgery, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
MicroRNAs (miRs) are small, noncoding RNA molecules with important regulatory functions whose role in regulating natural killer (NK) cell biology is not well defined. Here, we show that miR-155 is synergistically induced in primary human NK cells after costimulation with IL-12 and IL-18, or with IL-12 and CD16 clustering. Over-expression of miR-155 enhanced induction of IFN-γ by IL-12 and IL-18 or CD16 stimulation, whereas knockdown of miR-155 or its disruption suppressed IFN-γ induction in monokine and/or CD16-stimulated NK cells. These effects on the regulation of NK cell IFN-γ expression were found to be mediated at least in part via miR-155's direct effects on the inositol phosphatase SHIP1. Consistent with this, we observed that modulation of miR-155 overrides IL-12 and IL-18-mediated regulation of SHIP1 expression in NK cells. Collectively, our data indicate that miR-155 expression is regulated by stimuli that strongly induce IFN-γ in NK cells such as IL-12, IL-18, and CD16 activation, and that miR-155 functions as a positive regulator of IFN-γ production in human NK cells, at least in part via down-regulating SHIP1. These findings may have clinical relevance for targeting miR-155 in neoplastic disease.
Collapse
|
49
|
Antibody-dependent anti-cytomegalovirus activity of human γδ T cells expressing CD16 (FcγRIIIa). Blood 2012; 119:1418-27. [DOI: 10.1182/blood-2011-06-363655] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Human cytomegalovirus (HCMV) infection is an important cause of morbidity and mortality in transplant recipients. Long-term protective immunity against HCMV requires both sustained specific T-cell response and neutralizing IgG production, but the interplay between these effector arms remains poorly defined. We previously demonstrated that γδ T cells play a substantial role as anti-HCMV T-cell effectors. The observation that CD16 (FcγRIIIA) was specifically expressed by the majority of HCMV-induced γδ T cells prompted us to investigate their cooperation with anti-HCMV IgG. We found that CD16 could stimulate γδ T cells independently of T-cell receptor (TCR) engagement and provide them with an intrinsic antibody-dependent cell-mediated cytotoxic (ADCC) potential. Although CD16+γδ T cells did not mediate ADCC against HCMV-infected cells, in accordance with the low level of anti-HCMV IgGs recognizing infected cells, they produced IFNγ when incubated with IgG-opsonized virions. This CD16-induced IFNγ production was greatly enhanced by IL12 and IFNα, 2 cytokines produced during HCMV infection, and conferred to γδ T cells the ability to inhibit HCMV multiplication in vitro. Taken together, these data identify a new antiviral function for γδ T cells through cooperation with anti-HCMV IgG that could contribute to surveillance of HCMV reactivation in transplant recipients.
Collapse
|
50
|
Meng X, Guo A, Gong W, Jia W, Luo X, Zhai J, Dou Y, Cai X. Molecular characterization, tissue distribution and expression analysis of interleukin-12 receptor β2 chain in sheep. Gene 2012; 499:124-9. [PMID: 22265841 DOI: 10.1016/j.gene.2011.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/14/2011] [Accepted: 12/06/2011] [Indexed: 10/14/2022]
Abstract
Ovine β2 subunit of the interleukin (IL)-12 receptor (IL-12Rβ2) was cloned from mRNA preparation of mitogen-activated peripheral blood mononuclear cells (PBMCs). The complete coding sequence for ovine IL-12 Rβ2 was found to be 2586 nucleotides in length encoding 862-amino-acid residue protein. It showed 96.4% homology at the nucleotide level and 94.1% homology at the amino acid level with bovine IL-12 Rβ2. The ovine IL-12 Rβ2 subunit shares common structural and functional elements with their counterparts from the other species. Phylogenetic tree showed that ovine IL-12Rβ2 was clustered into the Artiodactyla group, together with those of cattle and pig, which was distinct from the other groups. Real-time RT-PCR was used to investigate expression of the IL-12Rβ2 in different tissues of sheep in order to determine the characterization of this receptor in tissue. Expression analysis showed that IL-12Rβ2 mRNA expression was detected at all the detected tissues with the exception of thymus.
Collapse
Affiliation(s)
- Xuelian Meng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, China
| | | | | | | | | | | | | | | |
Collapse
|