51
|
Chaves P, Zriwil A, Wittmann L, Boukarabila H, Peitzsch C, Jacobsen SEW, Sitnicka E. Loss of Canonical Notch Signaling Affects Multiple Steps in NK Cell Development in Mice. THE JOURNAL OF IMMUNOLOGY 2018; 201:3307-3319. [PMID: 30366956 DOI: 10.4049/jimmunol.1701675] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 09/27/2018] [Indexed: 11/19/2022]
Abstract
Within the hematopoietic system, the Notch pathway is critical for promoting thymic T cell development and suppressing the B and myeloid lineage fates; however, its impact on NK lymphopoiesis is less understood. To study the role of Notch during NK cell development in vivo, we investigated different NK cell compartments and function in Rbp-Jkfl/flVav-Cretg/+ mice, in which Rbp-Jk, the major transcriptional effector of canonical Notch signaling, was specifically deleted in all hematopoietic cells. Peripheral conventional cytotoxic NK cells in Rbp-Jk-deleted mice were significantly reduced and had an activated phenotype. Furthermore, the pool of early NK cell progenitors in the bone marrow was decreased, whereas immature NK cells were increased, leading to a block in NK cell maturation. These changes were cell intrinsic as the hematopoietic chimeras generated after transplantation of Rbp-Jk-deficient bone marrow cells had the same NK cell phenotype as the Rbp-Jk-deleted donor mice, whereas the wild-type competitors did not. The expression of several crucial NK cell regulatory pathways was significantly altered after Rbp-Jk deletion. Together, these results demonstrate the involvement of canonical Notch signaling in regulation of multiple stages of NK cell development.
Collapse
Affiliation(s)
- Patricia Chaves
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden.,Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| | - Alya Zriwil
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden.,Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| | - Lilian Wittmann
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden.,Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| | - Hanane Boukarabila
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Claudia Peitzsch
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden
| | - Sten Eirik W Jacobsen
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom.,MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom.,Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 86 Stockholm, Sweden; and.,Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ewa Sitnicka
- Lund Research Center for Stem Cell Biology and Cell Therapy, Lund University, 221 84 Lund, Sweden; .,Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
52
|
Li XY, Das I, Lepletier A, Addala V, Bald T, Stannard K, Barkauskas D, Liu J, Aguilera AR, Takeda K, Braun M, Nakamura K, Jacquelin S, Lane SW, Teng MW, Dougall WC, Smyth MJ. CD155 loss enhances tumor suppression via combined host and tumor-intrinsic mechanisms. J Clin Invest 2018; 128:2613-2625. [PMID: 29757192 DOI: 10.1172/jci98769] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/16/2018] [Indexed: 12/20/2022] Open
Abstract
Critical immune-suppressive pathways beyond programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) require greater attention. Nectins and nectin-like molecules might be promising targets for immunotherapy, since they play critical roles in cell proliferation and migration and exert immunomodulatory functions in pathophysiological conditions. Here, we show CD155 expression in both malignant cells and tumor-infiltrating myeloid cells in humans and mice. Cd155-/- mice displayed reduced tumor growth and metastasis via DNAM-1 upregulation and enhanced effector function of CD8+ T and NK cells, respectively. CD155-deleted tumor cells also displayed slower tumor growth and reduced metastases, demonstrating the importance of a tumor-intrinsic role of CD155. CD155 absence on host and tumor cells exerted an even greater inhibition of tumor growth and metastasis. Blockade of PD-1 or both PD-1 and CTLA4 was more effective in settings in which CD155 was limiting, suggesting the clinical potential of cotargeting PD-L1 and CD155 function.
Collapse
Affiliation(s)
- Xian-Yang Li
- Immunology in Cancer and Infection Laboratory and
| | - Indrajit Das
- Immunology in Cancer and Infection Laboratory and
| | | | - Venkateswar Addala
- Medical Genomics, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Tobias Bald
- Immunology in Cancer and Infection Laboratory and
| | | | | | - Jing Liu
- Immunology in Cancer and Infection Laboratory and
| | | | - Kazuyoshi Takeda
- Division of Cell Biology, Biomedical Research Center and Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | | | | | - Sebastien Jacquelin
- Gordon and Jessie Gilmour Leukaemia Research Laboratory, Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Steven W Lane
- Gordon and Jessie Gilmour Leukaemia Research Laboratory, Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,The Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Michele Wl Teng
- School of Medicine, The University of Queensland, Herston, Queensland, Australia.,Cancer Immunoregulation and Immunotherapy and
| | - William C Dougall
- Immunology in Cancer and Infection Laboratory and.,Immuno-oncology Discovery, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory and.,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
53
|
Wang M, Bu J, Zhou M, Sido J, Lin Y, Liu G, Lin Q, Xu X, Leavenworth JW, Shen E. CD8 +T cells expressing both PD-1 and TIGIT but not CD226 are dysfunctional in acute myeloid leukemia (AML) patients. Clin Immunol 2018; 190:64-73. [PMID: 28893624 DOI: 10.1016/j.clim.2017.08.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/16/2017] [Accepted: 08/24/2017] [Indexed: 01/07/2023]
Abstract
Acute myeloid leukemia (AML) is one of the most common types of leukemia among adults with an overall poor prognosis and very limited treatment management. Immune checkpoint blockade of PD-1 alone or combined with other immune checkpoint blockade has gained impressive results in murine AML models by improving anti-leukemia CD8+T cell function, which has greatly promoted the strategy to utilize combined immune checkpoint inhibitors to treat AML patients. However, the expression profiles of these immune checkpoint receptors, such as co-inhibitory receptors PD-1 and TIGIT and co-stimulatory receptor CD226, in T cells from AML patients have not been clearly defined. Here we have defined subsets of CD8+ and CD4+ T cells in the peripheral blood (PB) from newly diagnosed AML patients and healthy controls (HCs). We have observed increased frequencies of PD-1- and TIGIT- expressing CD8+ T cells but decreased occurrence of CD226-expressing CD8+T cells in AML patients. Further analysis of these CD8+ T cells revealed a unique CD8+ T cell subset that expressed PD-1 and TIGIT but displayed lower levels of CD226 was associated with failure to achieve remission after induction chemotherapy and FLT3-ITD mutations which predict poor clinical prognosis in AML patients. Importantly, these PD-1+TIGIT+CD226-CD8+T cells are dysfunctional with lower expression of intracellular IFN-γ and TNF-α than their counterparts in HCs. Therefore, our studies revealed that an increased frequency of a unique CD8+ T cell subset, PD-1+TIGIT+CD226-CD8+T cells, is associated with CD8+T cell dysfunction and poor clinical prognosis of AML patients, which may reveal critical diagnostic or prognostic biomarkers and direct more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Mengjie Wang
- Department of Pathogenic Biology and Immunology, Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 510182, China
| | - Jin Bu
- Editorial Department of Journals of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Maohua Zhou
- Department of Laboratory Medicine, Guangdong General Hospital, Academy of Medical Sciences, Guangzhou 510080, China
| | - Jessica Sido
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA 02115, USA; Department of Microbiology & Immunobiology, Division of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Yu Lin
- Shenzhen Withsum Technology Limited, Shenzhen 518031, China
| | - Guanfang Liu
- Department of Pathogenic Biology and Immunology, Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 510182, China
| | - Qiwen Lin
- Guangzhou Blood Center, Guangzhou 510095, China
| | - Xiuzhang Xu
- Guangzhou Blood Center, Guangzhou 510095, China
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Erxia Shen
- Department of Pathogenic Biology and Immunology, Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 510182, China; Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
54
|
Altered expression of CD226 and CD96 on natural killer cells in patients with pancreatic cancer. Oncotarget 2018; 7:66586-66594. [PMID: 27626490 PMCID: PMC5341822 DOI: 10.18632/oncotarget.11953] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 09/02/2016] [Indexed: 01/05/2023] Open
Abstract
The progression of pancreatic cancer (PC) is significantly associated with tumor immune escape, which may be associated with nature killer (NK) cell dysfunction. CD226, CD96, and TIGIT, which share the ligand CD155, play important roles in the regulation of NK cell function. The present study was conducted to investigate the roles of these molecules in NK cells from PC patients. Expression of these molecules on NK cells was detected from samples of 92 pancreatic cancer patients and 40 healthy controls. The expression of CD155 was also evaluated by immunohistochemistry in 88 pancreatic cancer tissues. The percentage of CD226+ and CD96+ NK cells was significantly lower in PC patients than in the healthy controls; however, the mean fluorescence intensity of CD226 and CD96 was not significantly different between the two groups. TIGIT expression on NK cells from PC patients was similar to that in the healthy controls. Additionally, the expression of CD226 was positively correlated with CD96. Further analysis demonstrated that the decrease in the percentage of CD226+ and CD96+ NK cells was associated with tumor histological grade and lymph node metastasis. Moreover, the CD155 levels in PC tissues were significantly higher than those in adjacent tissues. Our results suggest that a lower percentage of CD226+ and CD96+ NK cells may contribute to tumor immune escape in PC patients; moreover, the use of NK cells with high CD226 and CD96 expression to treat PC cells with high CD155 expression may have potential and should be explored in the future.
Collapse
|
55
|
Mehta RS, Randolph B, Daher M, Rezvani K. NK cell therapy for hematologic malignancies. Int J Hematol 2018; 107:262-270. [PMID: 29383623 DOI: 10.1007/s12185-018-2407-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/22/2018] [Indexed: 12/17/2022]
Abstract
Natural killer (NK) cells are part of the innate immune system and represent the first line of defense against infections and tumors. In contrast to T cells, NK cells do not require prior antigen sensitization to induce cytotoxicity and do not cause graft-versus-host disease. These, along with other advantages, make NK cells an attractive candidate for adoptive cellular therapy. Herein, we describe the mechanisms of NK cell cytotoxicity, which is governed by an intricate balance between various activating and inhibitory receptors, including the killer cell immunoglobulin-like receptors (KIRs). We illustrate the advantages of NK alloreactivity as demonstrated in various types of hematopoietic stem cell transplants (HSCT), such as haploidentical, human leukocyte antigen-matched related or unrelated donor and umbilical cord blood transplant. We elaborate on different models used to predict NK cell alloreactivity in these studies, which are either based on the absence of the ligands for inhibitory KIRs, presence of activating NK cell receptors and KIR genes content in donors, or a combination of these. We will review clinical studies demonstrating anti-tumor efficacy of NK cells used either as a stand-alone immunotherapy or as an adjunct to HSCT and novel genetic engineering strategies to improve the anti-tumor activity of NK cells.
Collapse
Affiliation(s)
- Rohtesh S Mehta
- Department of Stem Cell Transplant and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Unit 0423, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Brion Randolph
- Department of Stem Cell Transplant and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Unit 0423, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - May Daher
- Department of Stem Cell Transplant and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Unit 0423, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplant and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Unit 0423, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| |
Collapse
|
56
|
Epstein-Barr Virus Induces Adhesion Receptor CD226 (DNAM-1) Expression during Primary B-Cell Transformation into Lymphoblastoid Cell Lines. mSphere 2017; 2:mSphere00305-17. [PMID: 29202043 PMCID: PMC5705804 DOI: 10.1128/msphere.00305-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/16/2017] [Indexed: 12/14/2022] Open
Abstract
Epstein-Barr virus (EBV), an oncogenic herpesvirus, infects and transforms primary B cells into immortal lymphoblastoid cell lines (LCLs), providing a model for EBV-mediated tumorigenesis. EBV transformation stimulates robust homotypic aggregation, indicating that EBV induces molecules that mediate cell-cell adhesion. We report that EBV potently induced expression of the adhesion molecule CD226, which is not normally expressed on B cells. We found that early after infection of primary B cells, EBV promoted an increase in CD226 mRNA and protein expression. CD226 levels increased further from early proliferating EBV-positive B cells to LCLs. We found that CD226 expression on B cells was independent of B-cell activation as CpG DNA failed to induce CD226 to the extent of EBV infection. CD226 expression was high in EBV-infected B cells expressing the latency III growth program, but low in EBV-negative and EBV latency I-infected B-lymphoma cell lines. We validated this correlation by demonstrating that the latency III characteristic EBV NF-κB activator, latent membrane protein 1 (LMP1), was sufficient for CD226 upregulation and that CD226 was more highly expressed in lymphomas with increased NF-κB activity. Finally, we found that CD226 was not important for LCL steady-state growth, survival in response to apoptotic stress, homotypic aggregation, or adhesion to activated endothelial cells. These findings collectively suggest that EBV induces expression of a cell adhesion molecule on primary B cells that may play a role in the tumor microenvironment of EBV-associated B-cell malignancies or facilitate adhesion in the establishment of latency in vivo. IMPORTANCE Epstein-Barr virus (EBV) is a common human herpesvirus that establishes latency in B cells. While EBV infection is asymptomatic for most individuals, immune-suppressed individuals are at significantly higher risk of a form of EBV latent infection in which infected B cells are reactivated, grow unchecked, and generate lymphomas. This form of latency is modeled in the laboratory by infecting B cells from the blood of normal human donors in vitro. In this model, we identified a protein called CD226 that is induced by EBV but is not normally expressed on B cells. Rather, it is known to play a role in aggregation and survival signaling of non-B cells in the immune system. Cultures of EBV-infected cells adhere to one another in "clumps," and while the proteins that are responsible for this cellular aggregation are not fully understood, we hypothesized that this form of cellular aggregation may provide a survival advantage. In this article, we characterize the mechanism by which EBV induces this protein and its expression on lymphoma tissue and cell lines and characterize EBV-infected cell lines in which CD226 has been knocked out.
Collapse
|
57
|
Zhang R, Zeng H, Zhang Y, Chen K, Zhang C, Song C, Fang L, Xu Z, Yang K, Jin B, Wang Q, Chen L. CD226 ligation protects against EAE by promoting IL-10 expression via regulation of CD4+ T cell differentiation. Oncotarget 2017; 7:19251-64. [PMID: 26942885 PMCID: PMC4991380 DOI: 10.18632/oncotarget.7834] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/29/2016] [Indexed: 11/25/2022] Open
Abstract
Treatment targeting CD226 can ameliorate experimental autoimmune encephalomyelitis (EAE), the widely accepted model of MS. However, the mechanisms still need to be elucidated. Here we showed that CD226 blockage by anti-CD226 blocking mAb LeoA1 efficiently promoted IL-10 production in human peripheral blood monocytes (PBMC) or in mixed lymphocyte culture (MLC) system, significantly induced the CD4+IL-10+ T cell differentiation while suppressing the generation of Th1 and Th17. Furthermore, CD226 pAb administration in vivo reduced the onset of EAE in mice by promoting IL-10 production and regulating T cell differentiation. Concomitantly, the onset and severity of EAE were reduced and the serum IL-10 expression levels were increased in CD226 knockout mice than that in control mice when both received EAE induction. These novel findings confirmed that CD226 played a pivotal role in mediating autoimmune diseases such as EAE. Furthermore, to our knowledge, we show for the first time that IL-10 is an important contributor in the inhibitory effects of CD226 ligation on EAE.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China.,State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Hanyu Zeng
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Yun Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Kun Chen
- Department of Neurobiology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Chunmei Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Chaojun Song
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Liang Fang
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Zhuwei Xu
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Kun Yang
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Qintao Wang
- State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Lihua Chen
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
58
|
Characteristics of NK cells from leukemic microenvironment in MLL-AF9 induced acute myeloid leukemia. Mol Immunol 2017; 93:68-78. [PMID: 29154208 DOI: 10.1016/j.molimm.2017.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 10/18/2017] [Accepted: 11/07/2017] [Indexed: 01/27/2023]
Abstract
NK cells are indispensable components of tissue microenvironment and play vital in both innate and adaptive immunity. The activation and function of NK cells are affected by tumor microenvironments. NK cells are also important players in leukemic microenvironment. However, their characteristics in leukemic microenvironment, including maturation status, phenotype, subpopulations and functional roles especially immunoregulatory potential, have not been well established. Here, we studied these characteristics of NK cells in MLL-AF9 induced mouse acute myeloid leukemia (AML) model. Increase of more mature NK cells were detected in the AML spleen. Splenic AML microenvironment promoted NK cell activation in early and middle stages of leukemia. Cytotoxicity molecules and cytokines were up-regulated in activated NK cells. Furthermore, NK cells from AML microenvironment regulated T cell function, not only by maintaining the activation of CD4+ and promoting the degranulation of cytotoxic CD8+ T cells but also by influencing the differentiation of CD4+ T cells. Moreover, two NK cell subpopulations marked by DNAM-1 (CD226) had distinct cytokine expression patterns but similar regulatory effects on T cells. Collectively, these findings highlight the significance of immunoregulatory role of NK cells, and suggest novel therapeutic potential for leukemia by manipulating NK cell immunoregulatory activity.
Collapse
|
59
|
Goodier MR, Jonjić S, Riley EM, Juranić Lisnić V. CMV and natural killer cells: shaping the response to vaccination. Eur J Immunol 2017; 48:50-65. [PMID: 28960320 DOI: 10.1002/eji.201646762] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/14/2017] [Accepted: 09/22/2017] [Indexed: 12/14/2022]
Abstract
Cytomegaloviruses (CMVs) are highly prevalent, persistent human pathogens that not only evade but also shape our immune responses. Natural killer (NK) cells play an important role in the control of CMV and CMVs have in turn developed a plethora of immunoevasion mechanisms targeting NK cells. This complex interplay can leave a long-lasting imprint on the immune system in general and affect responses toward other pathogens and vaccines. This review aims to provide an overview of NK cell biology and development, the manipulation of NK cells by CMVs and the potential impact of these evasion strategies on responses to vaccination.
Collapse
Affiliation(s)
- Martin R Goodier
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Stipan Jonjić
- Department for Histology and Embryology and Center for Proteomics, Faculty of Medicine, University of Rijeka, Croatia
| | - Eleanor M Riley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Vanda Juranić Lisnić
- Department for Histology and Embryology and Center for Proteomics, Faculty of Medicine, University of Rijeka, Croatia
| |
Collapse
|
60
|
Lee J, Minden MD, Chen WC, Streck E, Chen B, Kang H, Arruda A, Ly D, Der SD, Kang S, Achita P, D'Souza C, Li Y, Childs RW, Dick JE, Zhang L. Allogeneic Human Double Negative T Cells as a Novel Immunotherapy for Acute Myeloid Leukemia and Its Underlying Mechanisms. Clin Cancer Res 2017; 24:370-382. [PMID: 29074605 DOI: 10.1158/1078-0432.ccr-17-2228] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/26/2017] [Accepted: 10/23/2017] [Indexed: 11/16/2022]
Abstract
Purpose: To explore the potential of ex vivo expanded healthy donor-derived allogeneic CD4 and CD8 double-negative cells (DNT) as a novel cellular immunotherapy for leukemia patients.Experimental Design: Clinical-grade DNTs from peripheral blood of healthy donors were expanded and their antileukemic activity and safety were examined using flow cytometry-based in vitro killing assays and xenograft models against AML patient blasts and healthy donor-derived hematopoietic cells. Mechanism of action was investigated using antibody-mediated blocking assays and recombinant protein treatment assays.Results: Expanded DNTs from healthy donors target a majority (36/46) of primary AML cells, including 9 chemotherapy-resistant patient samples in vitro, and significantly reduce the leukemia load in patient-derived xenograft models in a DNT donor-unrestricted manner. Importantly, allogeneic DNTs do not attack normal hematopoietic cells or affect hematopoietic stem/progenitor cell engraftment and differentiation, or cause xenogeneic GVHD in recipients. Mechanistically, DNTs express high levels of NKG2D and DNAM-1 that bind to cognate ligands preferentially expressed on AML cells. Upon recognition of AML cells, DNTs rapidly release IFNγ, which further increases NKG2D and DNAM-1 ligands' expression on AML cells. IFNγ pretreatment enhances the susceptibility of AML cells to DNT-mediated cytotoxicity, including primary AML samples that are otherwise resistant to DNTs, and the effect of IFNγ treatment is abrogated by NKG2D and DNAM-1-blocking antibodies.Conclusions: This study supports healthy donor-derived allogeneic DNTs as a therapy to treat patients with chemotherapy-resistant AML and also reveals interrelated roles of NKG2D, DNAM-1, and IFNγ in selective targeting of AML by DNTs. Clin Cancer Res; 24(2); 370-82. ©2017 AACR.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Biomarkers
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Drug Resistance, Neoplasm
- Graft vs Host Reaction/immunology
- Humans
- Immunophenotyping
- Immunotherapy, Adoptive/methods
- Interferon-gamma/biosynthesis
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Mice
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Transplantation, Homologous
Collapse
Affiliation(s)
- JongBok Lee
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Weihsu C Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Elena Streck
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Branson Chen
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Hyeonjeong Kang
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Dalam Ly
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Sandy D Der
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Sohyeong Kang
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Paulina Achita
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Cheryl D'Souza
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yueyang Li
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Richard W Childs
- National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Li Zhang
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada.
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
61
|
Schäfer C, Ascui G, Ribeiro CH, López M, Prados-Rosales R, González PA, Bueno SM, Riedel CA, Baena A, Kalergis AM, Carreño LJ. Innate immune cells for immunotherapy of autoimmune and cancer disorders. Int Rev Immunol 2017; 36:315-337. [PMID: 28933579 DOI: 10.1080/08830185.2017.1365145] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Modulation of the immune system has been widely targeted for the treatment of several immune-related diseases, such as autoimmune disorders and cancer, due to its crucial role in these pathologies. Current available therapies focus mainly on symptomatic treatment and are often associated with undesirable secondary effects. For several years, remission of disease and subsequently recovery of immune homeostasis has been a major goal for immunotherapy. Most current immunotherapeutic strategies are aimed to inhibit or potentiate directly the adaptive immune response by modulating antibody production and B cell memory, as well as the effector potential and memory of T cells. Although these immunomodulatory approaches have shown some success in the clinic with promising therapeutic potential, they have some limitations related to their effectiveness in disease models and clinical trials, as well as elevated costs. In the recent years, a renewed interest has emerged on targeting innate immune cells for immunotherapy, due to their high plasticity and ability to exert a potent and extremely rapid response, which can influence the outcome of the adaptive immune response. In this review, we discuss the immunomodulatory potential of several innate immune cells, as well as they use for immunotherapy, especially in autoimmunity and cancer.
Collapse
Affiliation(s)
- Carolina Schäfer
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Gabriel Ascui
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Carolina H Ribeiro
- b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Mercedes López
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Rafael Prados-Rosales
- c Centro de Investigaciones Cooperativas en Biociencias (CIC bioGUNE) , Bilbao , Spain
| | - Pablo A González
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,d Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Susan M Bueno
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,d Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Claudia A Riedel
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,e Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina , Universidad Andrés Bello , Santiago , Chile
| | - Andrés Baena
- f Departamento de Microbiología y Parasitología, Facultad de Medicina , Universidad de Antioquia , Medellín , Colombia
| | - Alexis M Kalergis
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,d Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile.,g Departamento de Endocrinología, Facultad de Medicina , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Leandro J Carreño
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| |
Collapse
|
62
|
Bowers JR, Readler JM, Sharma P, Excoffon KJDA. Poliovirus Receptor: More than a simple viral receptor. Virus Res 2017; 242:1-6. [PMID: 28870470 DOI: 10.1016/j.virusres.2017.09.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 12/27/2022]
Abstract
The human poliovirus receptor (PVR) is a cell surface protein with a multitude of functions in human biology. PVR was initially identified as the receptor for the human poliovirus and recent discoveries have given a greater insight into both its morphology and its function. Alternative splicing of the PVR gene results in a total of 4 alternatively spliced isoforms. Two of these isoforms lack a complete transmembrane domain and are considered soluble and block viral infection; the remaining two transmembrane isoforms differ only at their extreme C-terminal domains resulting in differential localization in epithelia and polarity of viral infection. In addition to its role as a receptor for the human poliovirus, several native biological functions have also been uncovered. PVR is an important cell adhesion protein and is involved in the transendothelial migration of leukocytes. Through its interactions with CD226 and TIGIT, transmembrane proteins found on leukocytes, PVR is a key regulator of the cell-mediated immune response. As PVR is differentially regulated in a broad spectrum of cancers, it has a strong potential for clinical use as a biomarker. PVR is also a possible target for novel cancer therapies. Utilizing its natural tropism for PVR, a genetically modified form of the live attenuated poliovirus vaccine is currently being tested for its ability to locate and destroy certain tumors. These recent studies emphasize the importance of PVR in human biology and demonstrate its utility beyond being a viral receptor protein.
Collapse
Affiliation(s)
- Jonathan R Bowers
- Department of Biological Sciences, Wright State University, Dayton, OH, 45435, United States
| | - James M Readler
- Department of Biological Sciences, Wright State University, Dayton, OH, 45435, United States
| | - Priyanka Sharma
- Department of Biological Sciences, Wright State University, Dayton, OH, 45435, United States
| | | |
Collapse
|
63
|
Sanchez-Correa B, Bergua JM, Pera A, Campos C, Arcos MJ, Bañas H, Duran E, Solana R, Tarazona R. In Vitro Culture with Interleukin-15 Leads to Expression of Activating Receptors and Recovery of Natural Killer Cell Function in Acute Myeloid Leukemia Patients. Front Immunol 2017; 8:931. [PMID: 28824651 PMCID: PMC5545593 DOI: 10.3389/fimmu.2017.00931] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 07/21/2017] [Indexed: 02/06/2023] Open
Abstract
Despite recent progress in the therapeutic approach of malignant hemopathies, their prognoses remain frequently poor. Immunotherapy could open a new window of great interest in this setting. Natural killer (NK) cells constitute an important area of research for hematologic malignancies, because this subpopulation is able to kill target cells spontaneously without previous sensitization, representing a novel tool in the treatment of them. Abnormal NK cytolytic function is observed in several hematological malignancies, including acute myeloid leukemia (AML) and myelodysplastic syndromes. Several mechanisms are involved in this abnormal function, such as decreased expression of activating receptors, increased expression of inhibitory receptors or defective expression of NK cell ligands on target cells. New immunotherapies are focused in identifying factors that could increase the expression of these activating receptors, to counteract inhibitory receptors expression, and therefore, to improve the NK cell cytotoxic capacities against tumor cells. In this work, we analyze the effect of interleukin (IL)-15 on the expression of NK cell-activating receptors that play a crucial role in the lysis of blasts from AML patients. Our results showed that IL-15 increased the surface expression of NKp30 on NK cells from healthy donors and AML patients with the consequent improvement of NK cell cytotoxicity. Besides, the upregulation of NKp30 induced by IL-15 is associated with an improvement of NK-mediated myeloid dendritic cells (DCs) maturation. NK cells cultured with IL-15 showed an upregulation of NKp30, which is associated with an increase anti-tumor activity and with an improved maturation of immature DCs. In our in vitro model, IL-15 exerted a great activating stimulus that could be used as novel immunotherapy in AML patients.
Collapse
Affiliation(s)
| | - Juan M Bergua
- Department of Haematology, Hospital San Pedro de Alcantara, Cáceres, Spain
| | - Alejandra Pera
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Red Espanola de Investigacion en Patologia Infecciosa (REIPI), Córdoba, Spain.,Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Carmen Campos
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Red Espanola de Investigacion en Patologia Infecciosa (REIPI), Córdoba, Spain
| | - Maria Jose Arcos
- Department of Haematology, Hospital San Pedro de Alcantara, Cáceres, Spain
| | - Helena Bañas
- Department of Haematology, Hospital San Pedro de Alcantara, Cáceres, Spain
| | - Esther Duran
- Histology and Pathology Unit, Faculty of Veterinary, University of Extremadura, Cáceres, Spain
| | - Rafael Solana
- Immunology Unit, University of Extremadura, Cáceres, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Red Espanola de Investigacion en Patologia Infecciosa (REIPI), Córdoba, Spain
| | | |
Collapse
|
64
|
Mahapatra S, Mace EM, Minard CG, Forbes LR, Vargas-Hernandez A, Duryea TK, Makedonas G, Banerjee PP, Shearer WT, Orange JS. High-resolution phenotyping identifies NK cell subsets that distinguish healthy children from adults. PLoS One 2017; 12:e0181134. [PMID: 28767726 PMCID: PMC5540415 DOI: 10.1371/journal.pone.0181134] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 06/27/2017] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells are critical in immune defense against infected, stressed or transformed cells. Their function is regulated by the heterogeneous expression of a wide array of surface receptors that shape its phenotypic diversity. Although NK cells develop in the bone marrow and secondary lymphoid tissues, substantive differentiation is apparent in the peripheral blood including known age-related variation. In order to gain greater insight into phenotypic and functional variation within peripheral blood NK cells across age groups, we used multi-parametric, polyfunctional flow cytometry to interrogate the NK cell variability in 20 healthy adults and 15 5-10, 11-15 and 16-20 year-old children. We found that the normative ranges in both adults and children displayed great inter-individual variation for most markers. While the expression of several receptors did not differ, among those that did, the majority of the differences existed between adults and the three pediatric groups, rather than among children of different ages. Interestingly, we also identified variation in the individual expression of some markers by sex and ethnicity. Combinatorial analysis of NK cell receptors revealed intermediate subsets between the CD56bright and CD56dim NK cells. Furthermore, on examining the NK cell diversity by age, adults were discovered to have the lowest developmental diversity. Thus, our findings identify previously unappreciated NK cell subsets potentially distinguishing children from adults and suggest functional correlates that may have relevance in age-specific host defense.
Collapse
Affiliation(s)
- Sanjana Mahapatra
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Emily M. Mace
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Charles G. Minard
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lisa R. Forbes
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alexander Vargas-Hernandez
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Teresa K. Duryea
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Residents’ Primary Care Group, Texas Children’s Hospital, Houston, Texas, United States of America
| | - George Makedonas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Pinaki P. Banerjee
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, United States of America
| | - William T. Shearer
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jordan S. Orange
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
65
|
Increased Soluble CD226 in Sera of Patients with Cutaneous T-Cell Lymphoma Mediates Cytotoxic Activity against Tumor Cells via CD155. J Invest Dermatol 2017; 137:1766-1773. [DOI: 10.1016/j.jid.2017.03.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 11/18/2022]
|
66
|
Nectin spot: a novel type of nectin-mediated cell adhesion apparatus. Biochem J 2017; 473:2691-715. [PMID: 27621480 DOI: 10.1042/bcj20160235] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/23/2016] [Indexed: 01/10/2023]
Abstract
Nectins are Ca(2+)-independent immunoglobulin (Ig) superfamily cell adhesion molecules constituting a family with four members, all of which have three Ig-like loops at their extracellular regions. Nectins play roles in the formation of a variety of cell-cell adhesion apparatuses. There are at least three types of nectin-mediated cell adhesions: afadin- and cadherin-dependent, afadin-dependent and cadherin-independent, and afadin- and cadherin-independent. In addition, nectins trans-interact with nectin-like molecules (Necls) with three Ig-like loops and other Ig-like molecules with one to three Ig-like loops. Furthermore, nectins and Necls cis-interact with membrane receptors and integrins, some of which are associated with the nectin-mediated cell adhesions, and play roles in the regulation of many cellular functions, such as cell polarization, movement, proliferation, differentiation, and survival, co-operatively with these cell surface proteins. The nectin-mediated cell adhesions are implicated in a variety of diseases, including genetic disorders, neural disorders, and cancers. Of the three types of nectin-mediated cell adhesions, the afadin- and cadherin-dependent apparatus has been most extensively investigated, but the examples of the third type of apparatus independent of afadin and cadherin are recently increasing and its morphological and functional properties have been well characterized. We review here recent advances in research on this type of nectin-mediated cell adhesion apparatus, which is named nectin spot.
Collapse
|
67
|
Kyaw T, Peter K, Li Y, Tipping P, Toh BH, Bobik A. Cytotoxic lymphocytes and atherosclerosis: significance, mechanisms and therapeutic challenges. Br J Pharmacol 2017; 174:3956-3972. [PMID: 28471481 DOI: 10.1111/bph.13845] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 04/02/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023] Open
Abstract
Cytotoxic lymphocytes encompass natural killer lymphocytes (cells) and cytotoxic T cells that include CD8+ T cells, natural killer (NK) T cells, γ, δ (γδ)-T cells and human CD4 + CD28- T cells. These cells play critical roles in inflammatory diseases and in controlling cancers and infections. Cytotoxic lymphocytes can be activated via a number of mechanisms that may involve dendritic cells, macrophages, cytokines or surface proteins on stressed cells. Upon activation, they secrete pro-inflammatory cytokines as well as anti-inflammatory cytokines, chemokines and cytotoxins to promote inflammation and the development of atherosclerotic lesions including vulnerable lesions, which are strongly implicated in myocardial infarctions and strokes. Here, we review the mechanisms that activate and regulate cytotoxic lymphocyte activity, including activating and inhibitory receptors, cytokines, chemokine receptors-chemokine systems utilized to home to inflamed lesions and cytotoxins and cytokines through which they affect other cells within lesions. We also examine their roles in human and mouse models of atherosclerosis and the mechanisms by which they exert their pathogenic effects. Finally, we discuss strategies for therapeutically targeting these cells to prevent the development of atherosclerotic lesions and vulnerable plaques and the challenge of developing highly targeted therapies that only minimally affect the body's immune system, avoiding the complications, such as increased susceptibility to infections, which are currently associated with many immunotherapies for autoimmune diseases. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Tin Kyaw
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Immunology, Monash University, Melbourne, Vic, Australia
| | - Yi Li
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Peter Tipping
- Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Ban-Hock Toh
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Alex Bobik
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Immunology, Monash University, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| |
Collapse
|
68
|
Oberschmidt O, Kloess S, Koehl U. Redirected Primary Human Chimeric Antigen Receptor Natural Killer Cells As an "Off-the-Shelf Immunotherapy" for Improvement in Cancer Treatment. Front Immunol 2017. [PMID: 28649246 PMCID: PMC5465249 DOI: 10.3389/fimmu.2017.00654] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Primary human natural killer (NK) cells recognize and subsequently eliminate virus infected cells, tumor cells, or other aberrant cells. However, cancer cells are able to develop tumor immune escape mechanisms to undermine this immune control. To overcome this obstacle, NK cells can be genetically modified to express chimeric antigen receptors (CARs) in order to improve specific recognition of cancer surface markers (e.g., CD19, CD20, and ErbB2). After target recognition, intracellular CAR domain signaling (CD3ζ, CD28, 4-1BB, and 2B4) leads to activation of PI3K or DNAX proteins (DAP10, DAP12) and finally to enhanced cytotoxicity, proliferation, and/or interferon γ release. This mini-review summarizes both the first preclinical trials with CAR-engineered primary human NK cells and the translational implications for “off-the-shelf immunotherapy” in cancer treatment. Signal transduction in NK cells as well as optimization of CAR signaling will be described, becoming more and more a focal point of interest in addition to redirected T cells. Finally, strategies to overcome off-target effects will be discussed in order to improve future clinical trials and to avoid attacking healthy tissues.
Collapse
Affiliation(s)
- Olaf Oberschmidt
- Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Stephan Kloess
- Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Ulrike Koehl
- Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| |
Collapse
|
69
|
Frazao A, Colombo M, Fourmentraux-Neves E, Messaoudene M, Rusakiewicz S, Zitvogel L, Vivier E, Vély F, Faure F, Dréno B, Benlalam H, Bouquet F, Savina A, Pasmant E, Toubert A, Avril MF, Caignard A. Shifting the Balance of Activating and Inhibitory Natural Killer Receptor Ligands on BRAFV600E Melanoma Lines with Vemurafenib. Cancer Immunol Res 2017; 5:582-593. [PMID: 28576831 DOI: 10.1158/2326-6066.cir-16-0380] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/10/2017] [Accepted: 05/25/2017] [Indexed: 11/16/2022]
Abstract
Over 60% of human melanoma tumors bear a mutation in the BRAF gene. The most frequent mutation is a substitution at codon 600 (V600E), leading to a constitutively active BRAF and overactivation of the MAPK pathway. Patients harboring mutated BRAF respond to kinase inhibitors such as vemurafenib. However, these responses are transient, and relapses are frequent. Melanoma cells are efficiently lysed by activated natural killer (NK) cells. Melanoma cells express several stress-induced ligands that are recognized by activating NK-cell receptors. We have investigated the effect of vemurafenib on the immunogenicity of seven BRAF-mutated melanoma cells to NK cells and on their growth and sensitivity to NK-cell-mediated lysis. We showed that vemurafenib treatment modulated expression of ligands for two activating NK receptors, increasing expression of B7-H6, a ligand for NKp30, and decreasing expression of MICA and ULBP2, ligands for NKG2D. Vemurafenib also increased expression of HLA class I and HLA-E molecules, likely leading to higher engagement of inhibitory receptors (KIRs and NKG2A, respectively), and decreased lysis of vemurafenib-treated melanoma cell lines by cytokine-activated NK cells. Finally, we showed that whereas batimastat (a broad-spectrum matrix metalloprotease inhibitor) increased cell surface ULBP2 by reducing its shedding, vemurafenib lowered soluble ULBP2, indicating that BRAF signal inhibition diminished expression of both cell-surface and soluble forms of NKG2D ligands. Vemurafenib, inhibiting BRAF signaling, shifted the balance of activatory and inhibitory NK ligands on melanoma cells and displayed immunoregulatory effects on NK-cell functional activities. Cancer Immunol Res; 5(7); 582-93. ©2017 AACR.
Collapse
Affiliation(s)
- Alexandra Frazao
- INSERM UMRS1160, Institut Universitaire d'Hématologie, Paris, France
| | - Marina Colombo
- INSERM UMRS1160, Institut Universitaire d'Hématologie, Paris, France
| | | | | | | | | | - Eric Vivier
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France.,Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, Service d'Immunologie, Marseille, France
| | - Frédéric Vély
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France.,Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, Service d'Immunologie, Marseille, France
| | | | - Brigitte Dréno
- UMR 892-CRCNA, Institut de Recherche Thérapeutique de l'Université de Nantes, Nantes, France
| | - Houssem Benlalam
- UMR 892-CRCNA, Institut de Recherche Thérapeutique de l'Université de Nantes, Nantes, France
| | | | | | - Eric Pasmant
- Service de Biochimie et Génétique Moléculaire, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Antoine Toubert
- INSERM UMRS1160, Institut Universitaire d'Hématologie, Paris, France
| | | | - Anne Caignard
- INSERM UMRS1160, Institut Universitaire d'Hématologie, Paris, France.
| |
Collapse
|
70
|
Veluchamy JP, Kok N, van der Vliet HJ, Verheul HMW, de Gruijl TD, Spanholtz J. The Rise of Allogeneic Natural Killer Cells As a Platform for Cancer Immunotherapy: Recent Innovations and Future Developments. Front Immunol 2017; 8:631. [PMID: 28620386 PMCID: PMC5450018 DOI: 10.3389/fimmu.2017.00631] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 05/12/2017] [Indexed: 12/20/2022] Open
Abstract
Natural killer (NK) cells are critical immune effector cells in the fight against cancer. As NK cells in cancer patients are highly dysfunctional and reduced in number, adoptive transfer of large numbers of cytolytic NK cells and their potential to induce relevant antitumor responses are widely explored in cancer immunotherapy. Early studies from autologous NK cells have failed to demonstrate significant clinical benefit. In this review, the clinical benefits of adoptively transferred allogeneic NK cells in a transplant and non-transplant setting are compared and discussed in the context of relevant NK cell platforms that are being developed and optimized by various biotech industries with a special focus on augmenting NK cell functions.
Collapse
Affiliation(s)
- John P Veluchamy
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands.,Glycostem Therapeutics, Oss, Netherlands
| | - Nina Kok
- Glycostem Therapeutics, Oss, Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
71
|
Beneficial in-vitro effects of interleukin-2, interleukin-12, and their combination on functional and receptor characteristics of natural killer cells in metastatic melanoma patients with normal serum lactate dehydrogenase levels. Melanoma Res 2016; 26:551-564. [DOI: 10.1097/cmr.0000000000000289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
72
|
Pignoloni B, Fionda C, Dell'Oste V, Luganini A, Cippitelli M, Zingoni A, Landolfo S, Gribaudo G, Santoni A, Cerboni C. Distinct Roles for Human Cytomegalovirus Immediate Early Proteins IE1 and IE2 in the Transcriptional Regulation of MICA and PVR/CD155 Expression. THE JOURNAL OF IMMUNOLOGY 2016; 197:4066-4078. [PMID: 27733551 DOI: 10.4049/jimmunol.1502527] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 09/09/2016] [Indexed: 12/22/2022]
Abstract
Elimination of virus-infected cells by cytotoxic lymphocytes is triggered by activating receptors, among which NKG2D and DNAM-1/CD226 play an important role. Their ligands, that is, MHC class I-related chain (MIC) A/B and UL16-binding proteins (ULBP)1-6 (NKG2D ligand), Nectin-2/CD112, and poliovirus receptor (PVR)/CD155 (DNAM-1 ligand), are often induced on virus-infected cells, although some viruses, including human CMV (HCMV), can block their expression. In this study, we report that infection of different cell types with laboratory or low-passage HCMV strains upregulated MICA, ULBP3, and PVR, with NKG2D and DNAM-1 playing a role in NK cell-mediated lysis of infected cells. Inhibition of viral DNA replication with phosphonoformic acid did not prevent ligand upregulation, thus indicating that early phases of HCMV infection are involved in ligand increase. Indeed, the major immediate early (IE) proteins IE1 and IE2 stimulated the expression of MICA and PVR, but not ULBP3. IE2 directly activated MICA promoter via its binding to an IE2-responsive element that we identified within the promoter and that is conserved among different alleles of MICA. Both IE proteins were instead required for PVR upregulation via a mechanism independent of IE DNA binding activity. Finally, inhibiting IE protein expression during HCMV infection confirmed their involvement in ligand increase. We also investigated the contribution of the DNA damage response, a pathway activated by HCMV and implicated in ligand regulation. However, silencing of ataxia telangiectasia mutated, ataxia telangiectasia and Rad3-related protein, and DNA-dependent protein kinase did not influence ligand expression. Overall, these data reveal that MICA and PVR are directly regulated by HCMV IE proteins, and this may be crucial for the onset of an early host antiviral response.
Collapse
Affiliation(s)
- Benedetta Pignoloni
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00162 Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00162 Rome, Italy
| | - Valentina Dell'Oste
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Anna Luganini
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; and
| | - Marco Cippitelli
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00162 Rome, Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00162 Rome, Italy
| | - Santo Landolfo
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Giorgio Gribaudo
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; and
| | - Angela Santoni
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00162 Rome, Italy; .,Mediterranean Neurological Institute-Neuromed, 86077 Pozzilli (Isernia), Italy
| | - Cristina Cerboni
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00162 Rome, Italy;
| |
Collapse
|
73
|
Peipp M, Derer S, Lohse S, Staudinger M, Klausz K, Valerius T, Gramatzki M, Kellner C. HER2-specific immunoligands engaging NKp30 or NKp80 trigger NK-cell-mediated lysis of tumor cells and enhance antibody-dependent cell-mediated cytotoxicity. Oncotarget 2016; 6:32075-88. [PMID: 26392331 PMCID: PMC4741660 DOI: 10.18632/oncotarget.5135] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 09/04/2015] [Indexed: 12/19/2022] Open
Abstract
NK cells detect tumors through activating surface receptors, which bind self-antigens that are frequently expressed upon malignant transformation. To increase the recognition of tumor cells, the extracellular domains of ligands of the activating NK cell receptors NKp30, NKp80 and DNAM-1 (i.e. B7-H6, AICL and PVR, respectively) were fused to a single-chain fragment variable (scFv) targeting the human epidermal growth factor receptor 2 (HER2), which is displayed by various solid tumors. The resulting immunoligands, designated B7-H6:HER2-scFv, AICL:HER2-scFv, and PVR:HER2-scFv, respectively, bound HER2 and the addressed NK cell receptor. However, whereas B7-H6:HER2-scFv and AICL:HER2-scFv triggered NK cells to kill HER2-positive breast cancer cells at nanomolar concentrations, PVR:HER2-scFv was not efficacious. Moreover, NK cell cytotoxicity was enhanced synergistically when B7-H6:HER2-scFv or AICL:HER2-scFv were applied in combination with another HER2-specific immunoligand engaging the stimulatory receptor NKG2D. In contrast, no improvements were achieved by combining B7-H6:HER2-scFv with AICL:HER2-scFv. Additionally, B7-H6:HER2-scFv and AICL:HER2-scFv enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) by the therapeutic antibodies trastuzumab and cetuximab synergistically, with B7-H6:HER2-scFv exhibiting a higher efficacy. In summary, antibody-derived proteins engaging NKp30 or NKp80 may represent attractive biologics to further enhance anti-tumor NK cell responses and may provide an innovative approach to sensitize tumor cells for antibody-based immunotherapy.
Collapse
Affiliation(s)
- Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Stefanie Derer
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Stefan Lohse
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Matthias Staudinger
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Katja Klausz
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Christian Kellner
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
74
|
Lenac Rovis T, Kucan Brlic P, Kaynan N, Juranic Lisnic V, Brizic I, Jordan S, Tomic A, Kvestak D, Babic M, Tsukerman P, Colonna M, Koszinowski U, Messerle M, Mandelboim O, Krmpotic A, Jonjic S. Inflammatory monocytes and NK cells play a crucial role in DNAM-1-dependent control of cytomegalovirus infection. J Exp Med 2016; 213:1835-50. [PMID: 27503073 PMCID: PMC4995080 DOI: 10.1084/jem.20151899] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 07/14/2016] [Indexed: 12/24/2022] Open
Abstract
Jonjic et al. show that inflammatory macrophages play an essential role in the control of murine CMV (MCMV) infection through a DNAM-1–PVR pathway. The poliovirus receptor (PVR) is a ubiquitously expressed glycoprotein involved in cellular adhesion and immune response. It engages the activating receptor DNAX accessory molecule (DNAM)-1, the inhibitory receptor TIGIT, and the CD96 receptor with both activating and inhibitory functions. Human cytomegalovirus (HCMV) down-regulates PVR expression, but the significance of this viral function in vivo remains unknown. Here, we demonstrate that mouse CMV (MCMV) also down-regulates the surface PVR. The m20.1 protein of MCMV retains PVR in the endoplasmic reticulum and promotes its degradation. A MCMV mutant lacking the PVR inhibitor was attenuated in normal mice but not in mice lacking DNAM-1. This attenuation was partially reversed by NK cell depletion, whereas the simultaneous depletion of mononuclear phagocytes abolished the virus control. This effect was associated with the increased expression of DNAM-1, whereas TIGIT and CD96 were absent on these cells. An increased level of proinflammatory cytokines in sera of mice infected with the virus lacking the m20.1 and an increased production of iNOS by inflammatory monocytes was observed. Blocking of CCL2 or the inhibition of iNOS significantly increased titer of the virus lacking m20.1. In this study, we have demonstrated that inflammatory monocytes, together with NK cells, are essential in the early control of CMV through the DNAM-1–PVR pathway.
Collapse
Affiliation(s)
- Tihana Lenac Rovis
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Paola Kucan Brlic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Noa Kaynan
- The Lautenberg Center for General and Tumor Immunology, The Hebrew University, The BioMedical Research Institute, Hadassah Medical School, Jerusalem 91120, Israel
| | - Vanda Juranic Lisnic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Ilija Brizic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Stefan Jordan
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Adriana Tomic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia Department of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Daria Kvestak
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Marina Babic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Pinchas Tsukerman
- The Lautenberg Center for General and Tumor Immunology, The Hebrew University, The BioMedical Research Institute, Hadassah Medical School, Jerusalem 91120, Israel
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Ulrich Koszinowski
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Martin Messerle
- Department of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, The Hebrew University, The BioMedical Research Institute, Hadassah Medical School, Jerusalem 91120, Israel
| | - Astrid Krmpotic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Stipan Jonjic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| |
Collapse
|
75
|
Kearney CJ, Ramsbottom KM, Voskoboinik I, Darcy PK, Oliaro J. Loss of DNAM-1 ligand expression by acute myeloid leukemia cells renders them resistant to NK cell killing. Oncoimmunology 2016; 5:e1196308. [PMID: 27622064 DOI: 10.1080/2162402x.2016.1196308] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 01/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is associated with poor natural killer (NK) cell function through aberrant expression of NK-cell-activating receptors and their ligands on tumor cells. These alterations are thought to promote formation of inhibitory NK-target cell synapses, in which killer cell degranulation is attenuated. Allogeneic stem cell transplantation can be effective in treating AML, through restoration of NK cell lytic activity. Similarly, agents that augment NK-cell-activating signals within the immunological synapse may provide some therapeutic benefit. However, the receptor-ligand interactions that critically dictate NK cell function in AML remain undefined. Here, we demonstrate that CD112/CD155 expression is required for DNAM-1-dependent killing of AML cells. Indeed, the low, or absent, expression of CD112/CD155 on multiple AML cell lines resulted in failure to stimulate optimal NK cell function. Importantly, isolated clones with low CD112/155 expression were resistant to NK cell killing while those expressing abundant levels of CD112/155 were highly susceptible. Attenuated NK cell killing in the absence of CD112/CD155 originated from decreased NK-target cell conjugation. Furthermore, we reveal by time-lapse microscopy, a significant increase in NK cell 'failed killing' in the absence of DNAM-1 ligands. Consequently, NK cells preferentially lysed ligand-expressing cells within heterogeneous populations, driving clonal selection of CD112/CD155-negative blasts upon NK cell attack. Taken together, we identify reduced CD155 expression as a major NK cell escape mechanism in AML and an opportunity for targeted immunotherapy.
Collapse
Affiliation(s)
- Conor J Kearney
- Immune Defense Laboratory, Cancer Immunology Division, The Peter MacCallum Cancer Center, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kelly M Ramsbottom
- Immune Defense Laboratory, Cancer Immunology Division, The Peter MacCallum Cancer Center , East Melbourne, Victoria, Australia
| | - Ilia Voskoboinik
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia; Killer Cell Biology Laboratory, Cancer Immunology Division, The Peter MacCallum Cancer Center, East Melbourne, Victoria, Australia
| | - Phillip K Darcy
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia; Immunotherapy Laboratory, Cancer Immunology Division, The Peter MacCallum Cancer Center, East Melbourne, Victoria, Australia
| | - Jane Oliaro
- Immune Defense Laboratory, Cancer Immunology Division, The Peter MacCallum Cancer Center, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
76
|
Braun M, Ress ML, Yoo YE, Scholz CJ, Eyrich M, Schlegel PG, Wölfl M. IL12-mediated sensitizing of T-cell receptor-dependent and -independent tumor cell killing. Oncoimmunology 2016; 5:e1188245. [PMID: 27622043 DOI: 10.1080/2162402x.2016.1188245] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/25/2016] [Accepted: 05/05/2016] [Indexed: 10/21/2022] Open
Abstract
Interleukin 12 (IL12) is a key inflammatory cytokine critically influencing Th1/Tc1-T-cell responses at the time of initial antigen encounter. Therefore, it may be exploited for cancer immunotherapy. Here, we investigated how IL12, and other inflammatory cytokines, shape effector functions of human T-cells. Using a defined culture system, we followed the gradual differentiation and function of antigen-specific CD8(+) T cells from their initial activation as naïve T cells through their expansion phase as early memory cells to full differentiation as clonally expanded effector T cells. The addition of IL12 8 days after the initial priming event initiated two mechanistically separate events: First, IL12 sensitized the T-cell receptor (TCR) for antigen-specific activation, leading to an approximately 10-fold increase in peptide sensitivity and, in consequence, enhanced tumor cell killing. Secondly, IL12 enabled TCR/HLA-independent activation and cytotoxicity: this "non-specific" effect was mediated by the NK cell receptor DNAM1 (CD226) and dependent on ligand expression of the target cells. This IL12 regulated, DNAM1-mediated killing is dependent on src-kinases as well as on PTPRC (CD45) activity. Thus, besides enhancing TCR-mediated activation, we here identified for the first time a second IL12 mediated mechanism leading to activation of a receptor-dependent killing pathway via DNAM1.
Collapse
Affiliation(s)
- Matthias Braun
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany; Else-Kröner Forschungskolleg for Interdisciplinary Translational Immunology, School of Medicine, University of Würzburg, Würzburg, Germany
| | - Marie L Ress
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg , Würzburg, Germany
| | - Young-Eun Yoo
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg , Würzburg, Germany
| | - Claus J Scholz
- Core Unit Systems Medicine, University of Würzburg , Würzburg, Germany
| | - Matthias Eyrich
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg , Würzburg, Germany
| | - Paul G Schlegel
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany; Clinical Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Matthias Wölfl
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany; Clinical Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| |
Collapse
|
77
|
Impaired NK-mediated regulation of T-cell activity in multiple sclerosis is reconstituted by IL-2 receptor modulation. Proc Natl Acad Sci U S A 2016; 113:E2973-82. [PMID: 27162345 DOI: 10.1073/pnas.1524924113] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system (CNS) resulting from a breakdown in peripheral immune tolerance. Although a beneficial role of natural killer (NK)-cell immune-regulatory function has been proposed, it still needs to be elucidated whether NK cells are functionally impaired as part of the disease. We observed NK cells in active MS lesions in close proximity to T cells. In accordance with a higher migratory capacity across the blood-brain barrier, CD56(bright) NK cells represent the major intrathecal NK-cell subset in both MS patients and healthy individuals. Investigating the peripheral blood and cerebrospinal fluid of MS patients treated with natalizumab revealed that transmigration of this subset depends on the α4β1 integrin very late antigen (VLA)-4. Although no MS-related changes in the migratory capacity of NK cells were observed, NK cells derived from patients with MS exhibit a reduced cytolytic activity in response to antigen-activated CD4(+) T cells. Defective NK-mediated immune regulation in MS is mainly attributable to a CD4(+) T-cell evasion caused by an impaired DNAX accessory molecule (DNAM)-1/CD155 interaction. Both the expression of the activating NK-cell receptor DNAM-1, a genetic alteration consistently found in MS-association studies, and up-regulation of the receptor's ligand CD155 on CD4(+) T cells are reduced in MS. Therapeutic immune modulation of IL-2 receptor restores impaired immune regulation in MS by increasing the proportion of CD155-expressing CD4(+) T cells and the cytolytic activity of NK cells.
Collapse
|
78
|
Cacalano NA. Regulation of Natural Killer Cell Function by STAT3. Front Immunol 2016; 7:128. [PMID: 27148255 PMCID: PMC4827001 DOI: 10.3389/fimmu.2016.00128] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 03/21/2016] [Indexed: 01/05/2023] Open
Abstract
Natural killer (NK) cells, key members of a distinct hematopoietic lineage, innate lymphoid cells, are not only critical effectors that mediate cytotoxicity toward tumor and virally infected cells but also regulate inflammation, antigen presentation, and the adaptive immune response. It has been shown that NK cells can regulate the development and activation of many other components of the immune response, such as dendritic cells, which in turn, modulate the function of NK cells in multiple synergistic feed back loops driven by cell–cell contact, and the secretion of cytokines and chemokines that control effector function and migration of cells to sites of immune activation. The signal transducer and activator of transcription (STAT)-3 is involved in driving almost all of the pathways that control NK cytolytic activity as well as the reciprocal regulatory interactions between NK cells and other components of the immune system. In the context of tumor immunology, NK cells are a first line of defense that eliminates pre-cancerous and transformed cells early in the process of carcinogenesis, through a mechanism of “immune surveillance.” Even after tumors become established, NK cells are critical components of anticancer immunity: dysfunctional NK cells are often found in the peripheral blood of cancer patients, and the lack of NK cells in the tumor microenvironment often correlates to poor prognosis. The pathways and soluble factors activated in tumor-associated NK cells, cancer cells, and regulatory myeloid cells, which determine the outcome of cancer immunity, are all critically regulated by STAT3. Using the tumor microenvironment as a paradigm, we present here an overview of the research that has revealed fundamental mechanisms through which STAT3 regulates all aspects of NK cell biology, including NK development, activation, target cell killing, and fine tuning of the innate and adaptive immune responses.
Collapse
Affiliation(s)
- Nicholas A Cacalano
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA , Los Angeles, CA , USA
| |
Collapse
|
79
|
Sanchez-Correa B, Campos C, Pera A, Bergua JM, Arcos MJ, Bañas H, Casado JG, Morgado S, Duran E, Solana R, Tarazona R. Natural killer cell immunosenescence in acute myeloid leukaemia patients: new targets for immunotherapeutic strategies? Cancer Immunol Immunother 2016; 65:453-63. [PMID: 26059279 PMCID: PMC11029066 DOI: 10.1007/s00262-015-1720-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/22/2015] [Indexed: 12/19/2022]
Abstract
Several age-associated changes in natural killer (NK) cell phenotype have been reported that contribute to the defective NK cell response observed in elderly patients. A remodelling of the NK cell compartment occurs in the elderly with a reduction in the output of immature CD56(bright) cells and an accumulation of highly differentiated CD56(dim) NK cells. Acute myeloid leukaemia (AML) is generally a disease of older adults. NK cells in AML patients show diminished expression of several activating receptors that contribute to impaired NK cell function and, in consequence, to AML blast escape from NK cell immunosurveillance. In AML patients, phenotypic changes in NK cells have been correlated with disease progression and survival. NK cell-based immunotherapy has emerged as a possibility for the treatment of AML patients. The understanding of age-associated alterations in NK cells is therefore necessary to define adequate therapeutic strategies in older AML patients.
Collapse
Affiliation(s)
| | - Carmen Campos
- Department of Immunology, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Avenida Menendez Pidal s/n, 14004, Córdoba, Spain
| | - Alejandra Pera
- Department of Immunology, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Avenida Menendez Pidal s/n, 14004, Córdoba, Spain
| | - Juan M Bergua
- Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain
| | - Maria Jose Arcos
- Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain
| | - Helena Bañas
- Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain
| | - Javier G Casado
- Immunology Unit, University of Extremadura, Cáceres, Spain
- Stem Cell Therapy Unit, Minimally Invasive Surgery Centre Jesus Uson, Cáceres, Spain
| | - Sara Morgado
- Immunology Unit, University of Extremadura, Cáceres, Spain
| | - Esther Duran
- Histology and Pathology Unit, Faculty of Veterinary, University of Extremadura, Cáceres, Spain
| | - Rafael Solana
- Department of Immunology, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Avenida Menendez Pidal s/n, 14004, Córdoba, Spain.
| | | |
Collapse
|
80
|
Zhang B, Zhao W, Li H, Chen Y, Tian H, Li L, Zhang L, Gao C, Zheng J. Immunoreceptor TIGIT inhibits the cytotoxicity of human cytokine-induced killer cells by interacting with CD155. Cancer Immunol Immunother 2016; 65:305-14. [PMID: 26842126 PMCID: PMC11029225 DOI: 10.1007/s00262-016-1799-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/19/2016] [Indexed: 12/22/2022]
Abstract
T cell Ig and ITIM domain (TIGIT) is a newly identified inhibitory receptor expressed on T and natural killer (NK) cells. Cytokine-induced killer (CIK) cells express CD3 and CD56 molecules, and share functional properties with both NK and T cells. However, it remains unknown whether TIGIT is expressed in CIK cells. Here, we show that TIGIT is expressed by CIK cells and interacts with CD155. By blocking TIGIT using an anti-TIGIT functional antibody, we demonstrate that CIK cells display increased proliferation; higher cytotoxic targeting of tumor cells expressing CD155; and higher expression of interferon-γ (IFN-γ), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). Furthermore, increases in IFN-γ and cytotoxicity by blockade of TIGIT were reduced by blocking DNAX accessory molecule-1 (DNAM-1) signaling, implying that TIGIT exerts immunosuppressive effects by competing with DNAM-1 for the same ligand, CD155. Our results provide evidence that blockade of TIGIT may be a novel strategy to improve the cytotoxic activity of CIK cells.
Collapse
Affiliation(s)
- Baofu Zhang
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Weina Zhao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Huizhong Li
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Yuanyuan Chen
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Hui Tian
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Liantao Li
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Longzhen Zhang
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Chao Gao
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China.
| | - Junnian Zheng
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
81
|
Chester C, Fritsch K, Kohrt HE. Natural Killer Cell Immunomodulation: Targeting Activating, Inhibitory, and Co-stimulatory Receptor Signaling for Cancer Immunotherapy. Front Immunol 2015; 6:601. [PMID: 26697006 PMCID: PMC4667030 DOI: 10.3389/fimmu.2015.00601] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 11/09/2015] [Indexed: 01/11/2023] Open
Abstract
There is compelling clinical and experimental evidence to suggest that natural killer (NK) cells play a critical role in the recognition and eradication of tumors. Efforts at using NK cells as antitumor agents began over two decades ago, but recent advances in elucidating NK cell biology have accelerated the development of NK cell-targeting therapeutics. NK cell activation and the triggering of effector functions is governed by a complex set of activating and inhibitory receptors. In the early phases of cancer immune surveillance, NK cells directly identify and lyse cancer cells. Nascent transformed cells elicit NK cell activation and are eliminated. However, as tumors progress, cancerous cells develop immunosuppressive mechanisms that circumvent NK cell-mediated killing, allowing for tumor escape and proliferation. Therapeutic intervention aims to reverse tumor-induced NK cell suppression and sustain NK cells’ tumorlytic capacities. Here, we review tumor–NK cell interactions, discuss the mechanisms by which NK cells generate an antitumor immune response, and discuss NK cell-based therapeutic strategies targeting activating, inhibitory, and co-stimulatory receptors.
Collapse
Affiliation(s)
- Cariad Chester
- Division of Oncology, Department of Medicine, Stanford University , Stanford, CA , USA ; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine , Stanford, CA , USA
| | - Katherine Fritsch
- Division of Oncology, Department of Medicine, Stanford University , Stanford, CA , USA
| | - Holbrook E Kohrt
- Division of Oncology, Department of Medicine, Stanford University , Stanford, CA , USA
| |
Collapse
|
82
|
|
83
|
Zhang Z, Wu N, Lu Y, Davidson D, Colonna M, Veillette A. DNAM-1 controls NK cell activation via an ITT-like motif. J Exp Med 2015; 212:2165-82. [PMID: 26552706 PMCID: PMC4647266 DOI: 10.1084/jem.20150792] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/25/2015] [Indexed: 11/28/2022] Open
Abstract
DNAM-1–mediated active biochemical signals initiated by a conserved ITT-like motif are essential for its capacity to enhance NK cell cytotoxicity and cytokine production. Enhancement of these signals may underlie the therapeutic impact of blocking anti-TIGIT antibodies for treatment of cancer and viral infections. DNAM-1 (CD226) is an activating receptor expressed on natural killer (NK) cells, CD8+ T cells, and other immune cells. Upon recognition of its ligands, CD155 and CD112, DNAM-1 promotes NK cell–mediated elimination of transformed and virus-infected cells. It also has a key role in expansion and maintenance of virus-specific memory NK cells. Herein, the mechanism by which DNAM-1 controls NK cell–mediated cytotoxicity and cytokine production was elucidated. Cytotoxicity and cytokine production triggered by DNAM-1 were mediated via a conserved tyrosine- and asparagine-based motif in the cytoplasmic domain of DNAM-1. Upon phosphorylation by Src kinases, this motif enabled binding of DNAM-1 to adaptor Grb2, leading to activation of enzymes Vav-1, phosphatidylinositol 3′ kinase, and phospholipase C-γ1. It also promoted activation of kinases Erk and Akt, and calcium fluxes. Although, as reported, DNAM-1 promoted adhesion, this function was signal-independent and insufficient to promote cytotoxicity. DNAM-1 signaling was also required to enhance cytotoxicity, by increasing actin polymerization and granule polarization. We propose that DNAM-1 promotes NK cell activation via an immunoreceptor tyrosine tail (ITT)–like motif coupling DNAM-1 to Grb2 and other downstream effectors.
Collapse
Affiliation(s)
- Zhanguang Zhang
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec H2W 1R7, Canada Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Ning Wu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | - Yan Lu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | - Dominique Davidson
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec H2W 1R7, Canada Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada Department of Medicine, University of Montréal, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
84
|
Krasnova Y, Putz EM, Smyth MJ, Souza-Fonseca-Guimaraes F. Bench to bedside: NK cells and control of metastasis. Clin Immunol 2015; 177:50-59. [PMID: 26476139 DOI: 10.1016/j.clim.2015.10.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 10/08/2015] [Accepted: 10/14/2015] [Indexed: 12/22/2022]
Abstract
Natural killer (NK) cells play a critical role in host immune responses against tumor growth and metastasis. The numerous mechanisms used by NK cells to regulate and control cancer metastasis include interactions with tumor cells via specific receptors and ligands as well as direct cytotoxicity and cytokine-induced effector mechanisms. NK cells also play a role in tumor immunosurveillance and inhibition of metastases formation by recognition and killing of tumor cells. In this review, we provide an overview of the molecular mechanisms of NK cell responses against tumor metastases and discuss multiple strategies by which tumors evade NK cell-mediated surveillance. With an increasing understanding of the molecular mechanisms driving NK cell activity, there is a growing potential for the development of new cancer immunotherapies. Here we provide a historical background on NK cell-based therapies and discuss the implications of recent and ongoing clinical trials using novel NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Yelena Krasnova
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Medicine, University of Queensland, St Lucia, Queensland 4006, Australia
| | - Eva Maria Putz
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Medicine, University of Queensland, St Lucia, Queensland 4006, Australia
| | - Fernando Souza-Fonseca-Guimaraes
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Medicine, University of Queensland, St Lucia, Queensland 4006, Australia.
| |
Collapse
|
85
|
Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov 2015; 14:561-84. [PMID: 26228759 DOI: 10.1038/nrd4591] [Citation(s) in RCA: 996] [Impact Index Per Article: 99.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Targeting immune checkpoints such as programmed cell death protein 1 (PD1), programmed cell death 1 ligand 1 (PDL1) and cytotoxic T lymphocyte antigen 4 (CTLA4) has achieved noteworthy benefit in multiple cancers by blocking immunoinhibitory signals and enabling patients to produce an effective antitumour response. Inhibitors of CTLA4, PD1 or PDL1 administered as single agents have resulted in durable tumour regression in some patients, and combinations of PD1 and CTLA4 inhibitors may enhance antitumour benefit. Numerous additional immunomodulatory pathways as well as inhibitory factors expressed or secreted by myeloid and stromal cells in the tumour microenvironment are potential targets for synergizing with immune checkpoint blockade. Given the breadth of potential targets in the immune system, critical questions to address include which combinations should move forward in development and which patients will benefit from these treatments. This Review discusses the leading drug targets that are expressed on tumour cells and in the tumour microenvironment that allow enhancement of the antitumour immune response.
Collapse
Affiliation(s)
- Kathleen M Mahoney
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA. [2] Division of Haematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA. [3]
| | - Paul D Rennert
- 1] SugarCone Biotech, Holliston, Massachusetts 01746, USA. [2] Videre Biotherapeutics, Watertown, Massachusetts 02472, USA. [3]
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
86
|
Xiong P, Sang HW, Zhu M. Critical roles of co-activation receptor DNAX accessory molecule-1 in natural killer cell immunity. Immunology 2015; 146:369-78. [PMID: 26235210 DOI: 10.1111/imm.12516] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/28/2015] [Accepted: 07/28/2015] [Indexed: 12/19/2022] Open
Abstract
Natural killer (NK) cells, which can exert early and powerful anti-tumour and anti-viral responses, are important components of the innate immune system. DNAX accessory molecule-1 (DNAM-1) is an activating receptor molecule expressed on the surface of NK cells. Recent findings suggest that DNAM-1 is a critical regulator of NK cell biology. DNAM-1 is involved in NK cell education and differentiation, and also plays a pivotal role in the development of cancer, viral infections and immune-related diseases. However, tumours and viruses have developed multiple mechanisms to evade the immune system. They are able to impair DNAM-1 activity by targeting the DNAM-1 receptor-ligand system. We have reviewed the roles of DNAM-1, and its biological functions, with respect to NK cell biology and DNAM-1 chimeric antigen receptor-based immunotherapy.
Collapse
Affiliation(s)
- Peng Xiong
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Wei Sang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
87
|
Cantoni C, Grauwet K, Pietra G, Parodi M, Mingari MC, Maria AD, Favoreel H, Vitale M. Role of NK cells in immunotherapy and virotherapy of solid tumors. Immunotherapy 2015; 7:861-82. [PMID: 26314197 DOI: 10.2217/imt.15.53] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Although natural killer (NK) cells are endowed with powerful cytolytic activity against cancer cells, their role in different therapies against solid tumors has not yet been fully elucidated. Their interactions with various elements of the tumor microenvironment as well as their possible effects in contributing to and/or limiting oncolytic virotherapy render this potential immunotherapeutic tool still difficult to exploit at the bedside. Here, we will review the current literature with the aim of providing new hints to manage this powerful cell type in future innovative therapies, such as the use of NK cells in combination with new cytokines, specific mAbs (inducing ADCC), Tyr-Kinase inhibitors, immunomodulatory drugs and/or the design of oncolytic viruses aimed at optimizing the effect of NK cells in virotherapy.
Collapse
Affiliation(s)
- Claudia Cantoni
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,Istituto Giannina Gaslini, Genova, Italy
| | - Korneel Grauwet
- Laboratory of Immunology, Department of Virology, Parasitology & Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | - Gabriella Pietra
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy
| | - Monica Parodi
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy
| | - Andrea De Maria
- Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Herman Favoreel
- Laboratory of Immunology, Department of Virology, Parasitology & Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | | |
Collapse
|
88
|
Ansari AW, Ahmad F, Meyer-Olson D, Kamarulzaman A, Jacobs R, Schmidt RE. Natural killer cell heterogeneity: cellular dysfunction and significance in HIV-1 immuno-pathogenesis. Cell Mol Life Sci 2015; 72:3037-49. [PMID: 25939268 PMCID: PMC11113101 DOI: 10.1007/s00018-015-1911-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 04/17/2015] [Accepted: 04/20/2015] [Indexed: 11/28/2022]
Abstract
Natural killer (NK) cells are innate immune effectors that provide first line of defence against viruses. Human NK cells are heterogeneous in nature, and their functions rely on a dynamic balance between germ-line-encoded activating and inhibitory receptors. HIV-1 infection results in altered NK cell receptor repertoire and impaired effector functions including the ability to lyse virus-infected cells and secretion of antiviral cytokine IFN-γ. Over the last decade, additional NK cell subset-specific molecules have been identified, leading to emergence of a more complex cellular diversity than previously thought. Herein, we discuss NK cell subset redistribution, altered receptor repertoire and influence of interaction of polymorphic leucocyte antigen (HLA) and killer cell immunoglobulin-like receptors (KIR) on HIV-1 disease progression.
Collapse
Affiliation(s)
- A. Wahid Ansari
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Lambah Pantai, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, Faculty of Medicine, University of Malaya, Lambah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Fareed Ahmad
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | - Dirk Meyer-Olson
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | - Adeeba Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Lambah Pantai, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, Faculty of Medicine, University of Malaya, Lambah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Roland Jacobs
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | - Reinhold E. Schmidt
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| |
Collapse
|
89
|
The DNA damage response and immune signaling alliance: Is it good or bad? Nature decides when and where. Pharmacol Ther 2015; 154:36-56. [PMID: 26145166 DOI: 10.1016/j.pharmthera.2015.06.011] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 06/10/2015] [Indexed: 12/15/2022]
Abstract
The characteristic feature of healthy living organisms is the preservation of homeostasis. Compelling evidence highlight that the DNA damage response and repair (DDR/R) and immune response (ImmR) signaling networks work together favoring the harmonized function of (multi)cellular organisms. DNA and RNA viruses activate the DDR/R machinery in the host cells both directly and indirectly. Activation of DDR/R in turn favors the immunogenicity of the incipient cell. Hence, stimulation of DDR/R by exogenous or endogenous insults triggers innate and adaptive ImmR. The immunogenic properties of ionizing radiation, a prototypic DDR/R inducer, serve as suitable examples of how DDR/R stimulation alerts host immunity. Thus, critical cellular danger signals stimulate defense at the systemic level and vice versa. Disruption of DDR/R-ImmR cross talk compromises (multi)cellular integrity, leading to cell-cycle-related and immune defects. The emerging DDR/R-ImmR concept opens up a new avenue of therapeutic options, recalling the Hippocrates quote "everything in excess is opposed by nature."
Collapse
|
90
|
Battella S, Cox MC, Santoni A, Palmieri G. Natural killer (NK) cells and anti-tumor therapeutic mAb: unexplored interactions. J Leukoc Biol 2015; 99:87-96. [PMID: 26136506 DOI: 10.1189/jlb.5vmr0415-141r] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/16/2015] [Indexed: 12/11/2022] Open
Abstract
Tumor-targeting mAb are widely used in the treatment of a variety of solid and hematopoietic tumors and represent the first immunotherapeutic approach successfully arrived to the clinic. Nevertheless, the role of distinct immune mechanisms in contributing to their therapeutic efficacy is not completely understood and may vary depending on tumor- or antigen/antibody-dependent characteristics. Availability of next-generation, engineered, tumor-targeting mAb, optimized in their capability to recruit selected immune effectors, re-enforces the need for a deeper understanding of the mechanisms underlying anti-tumor mAb functionality. NK cells participate with a major role to innate anti-tumor responses, by exerting cytotoxic activity and producing a vast array of cytokines. As the CD16 (low-affinity FcγRIIIA)-activating receptor is expressed on the majority of NK cells, its effector functions can be ideally recruited against therapeutic mAb-opsonized tumor cells. The exact role of NK cells in determining therapeutic efficacy of tumor-targeting mAb is still unclear and much sought after. This knowledge will be instrumental to design innovative combination schemes with newly validated immunomodulatory agents. We will summarize what is known about the role of NK cells in therapeutic anti-tumor mAb therapy, with particular emphasis on RTX chimeric anti-CD20 mAb, the first one used in clinical practice for treating B cell malignancies.
Collapse
Affiliation(s)
- Simone Battella
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Maria Christina Cox
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Angela Santoni
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Gabriella Palmieri
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
91
|
Fuhrman CA, Yeh WI, Seay HR, Saikumar Lakshmi P, Chopra G, Zhang L, Perry DJ, McClymont SA, Yadav M, Lopez MC, Baker HV, Zhang Y, Li Y, Whitley M, von Schack D, Atkinson MA, Bluestone JA, Brusko TM. Divergent Phenotypes of Human Regulatory T Cells Expressing the Receptors TIGIT and CD226. THE JOURNAL OF IMMUNOLOGY 2015; 195:145-55. [PMID: 25994968 DOI: 10.4049/jimmunol.1402381] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 04/26/2015] [Indexed: 02/06/2023]
Abstract
Regulatory T cells (Tregs) play a central role in counteracting inflammation and autoimmunity. A more complete understanding of cellular heterogeneity and the potential for lineage plasticity in human Treg subsets may identify markers of disease pathogenesis and facilitate the development of optimized cellular therapeutics. To better elucidate human Treg subsets, we conducted direct transcriptional profiling of CD4(+)FOXP3(+)Helios(+) thymic-derived Tregs and CD4(+)FOXP3(+)Helios(-) T cells, followed by comparison with CD4(+)FOXP3(-)Helios(-) T conventional cells. These analyses revealed that the coinhibitory receptor T cell Ig and ITIM domain (TIGIT) was highly expressed on thymic-derived Tregs. TIGIT and the costimulatory factor CD226 bind the common ligand CD155. Thus, we analyzed the cellular distribution and suppressive activity of isolated subsets of CD4(+)CD25(+)CD127(lo/-) T cells expressing CD226 and/or TIGIT. We observed TIGIT is highly expressed and upregulated on Tregs after activation and in vitro expansion, and is associated with lineage stability and suppressive capacity. Conversely, the CD226(+)TIGIT(-) population was associated with reduced Treg purity and suppressive capacity after expansion, along with a marked increase in IL-10 and effector cytokine production. These studies provide additional markers to delineate functionally distinct Treg subsets that may help direct cellular therapies and provide important phenotypic markers for assessing the role of Tregs in health and disease.
Collapse
Affiliation(s)
- Christopher A Fuhrman
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Wen-I Yeh
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Howard R Seay
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Priya Saikumar Lakshmi
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Gaurav Chopra
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603
| | - Lin Zhang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Stephanie A McClymont
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603
| | - Mahesh Yadav
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603
| | - Maria-Cecilia Lopez
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco CA 94143
| | - Henry V Baker
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco CA 94143
| | - Ying Zhang
- Precision Medicine-Bioanalytical, Pfizer, Cambridge, MA 02139; and
| | - Yizheng Li
- Pfizer Research and Development Business Technologies, Cambridge, MA 02139
| | - Maryann Whitley
- Pfizer Research and Development Business Technologies, Cambridge, MA 02139
| | - David von Schack
- Precision Medicine-Bioanalytical, Pfizer, Cambridge, MA 02139; and
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Jeffrey A Bluestone
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco CA 94143
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610;
| |
Collapse
|
92
|
Guillerey C, Ferrari de Andrade L, Vuckovic S, Miles K, Ngiow SF, Yong MCR, Teng MWL, Colonna M, Ritchie DS, Chesi M, Bergsagel PL, Hill GR, Smyth MJ, Martinet L. Immunosurveillance and therapy of multiple myeloma are CD226 dependent. J Clin Invest 2015; 125:2077-89. [PMID: 25893601 DOI: 10.1172/jci77181] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 03/12/2015] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is an age-dependent hematological malignancy. Evaluation of immune interactions that drive MM relies on in vitro experiments that do not reflect the complex cellular stroma involved in MM pathogenesis. Here we used Vk*MYC transgenic mice, which spontaneously develop MM, and demonstrated that the immune system plays a critical role in the control of MM progression and the response to treatment. We monitored Vk*MYC mice that had been crossed with Cd226 mutant mice over a period of 3 years and found that CD226 limits spontaneous MM development. The CD226-dependent anti-myeloma immune response against transplanted Vk*MYC MM cells was mediated both by NK and CD8+ T cells through perforin and IFN-γ pathways. Moreover, CD226 expression was required for optimal antimyeloma efficacy of cyclophosphamide (CTX) and bortezomib (Btz), which are both standardly used to manage MM in patients. Activation of costimulatory receptor CD137 with mAb (4-1BB) exerted strong antimyeloma activity, while inhibition of coinhibitory receptors PD-1 and CTLA-4 had no effect. Taken together, the results of this study provide in vivo evidence that CD226 is important for MM immunosurveillance and indicate that specific immune components should be targeted for optimal MM treatment efficacy. As progressive immunosuppression associates with MM development, strategies aimed to increase immune functions may have important therapeutic implications in MM.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/physiology
- Antineoplastic Agents/therapeutic use
- Boronic Acids/therapeutic use
- Bortezomib
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen/antagonists & inhibitors
- Crosses, Genetic
- Cyclophosphamide/therapeutic use
- Disease Progression
- Genes, myc
- Genetic Predisposition to Disease
- Immunologic Surveillance/immunology
- Immunotherapy
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Killer Cells, Natural/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/immunology
- Neoplasm Proteins/deficiency
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Neoplasm Proteins/physiology
- Neoplasm Transplantation
- Pore Forming Cytotoxic Proteins/deficiency
- Pore Forming Cytotoxic Proteins/genetics
- Pore Forming Cytotoxic Proteins/physiology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Pyrazines/therapeutic use
- Receptors, Virus/deficiency
- Receptors, Virus/genetics
- Receptors, Virus/physiology
- Tumor Burden
- Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
Collapse
|
93
|
Decreased expression of NKG2D, NKp46, DNAM-1 receptors, and intracellular perforin and STAT-1 effector molecules in NK cells and their dim and bright subsets in metastatic melanoma patients. Melanoma Res 2015; 24:295-304. [PMID: 24769842 DOI: 10.1097/cmr.0000000000000072] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Although natural killer (NK) cells play an important antitumor role, melanoma cells may affect their effector functions. In this study, we analyzed the expression of various receptors and effector molecules in NK cells and their subsets in metastatic melanoma (MM) patients compared with healthy controls (HCs). In HC and MM patients, we analyzed NK cell activity using a chromium release assay and the expression of CD107a degranulation marker, activating NKG2D, NKp46, DNAM-1, and inhibitory CD158a and CD158b receptors, IL-12R beta 1, IL-12R beta 2, intracellular interferon (IFN)-γ, perforin, and STAT-1 in CD3-CD56+ NK cells, and cytotoxic CD3-CD56 and immunoregulatory CD3-CD56 subsets by flow cytometry. MM patients compared with HC not only had significantly decreased NK cell activity, lower expression of CD107a, and impaired IFN-γ production but also had decreased expression of activating NKG2D, NKp46, and DNAM-1 receptors, which was followed by lower expression of perforin, STAT-1, and both IL-12R subunits in NK cells. In MM patients only, there was a positive correlation between NKG2D expression and degranulation capacity, as well as IFN-γ production in NK cells. Analysis of the expression of various parameters of NK cell effector functions between MM patients with different localization of distant metastases showed that patients in the unfavorable M1c subclass had decreased expression of NKG2D and NKp46 on NK cells compared with patients in the M1a+b group. Downregulated NKG2D, NKp46, and DNAM-1 receptors associated with impaired NK cell effector function are important biomarkers of advanced disease with a poor prognosis in melanoma patients.
Collapse
|
94
|
Hsu JL, van den Boomen DJH, Tomasec P, Weekes MP, Antrobus R, Stanton RJ, Ruckova E, Sugrue D, Wilkie GS, Davison AJ, Wilkinson GWG, Lehner PJ. Plasma membrane profiling defines an expanded class of cell surface proteins selectively targeted for degradation by HCMV US2 in cooperation with UL141. PLoS Pathog 2015; 11:e1004811. [PMID: 25875600 PMCID: PMC4397069 DOI: 10.1371/journal.ppat.1004811] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 03/13/2015] [Indexed: 11/24/2022] Open
Abstract
Human cytomegalovirus (HCMV) US2, US3, US6 and US11 act in concert to prevent immune recognition of virally infected cells by CD8+ T-lymphocytes through downregulation of MHC class I molecules (MHC-I). Here we show that US2 function goes far beyond MHC-I degradation. A systematic proteomic study using Plasma Membrane Profiling revealed US2 was unique in downregulating additional cellular targets, including: five distinct integrin α-chains, CD112, the interleukin-12 receptor, PTPRJ and thrombomodulin. US2 recruited the cellular E3 ligase TRC8 to direct the proteasomal degradation of all its targets, reminiscent of its degradation of MHC-I. Whereas integrin α-chains were selectively degraded, their integrin β1 binding partner accumulated in the ER. Consequently integrin signaling, cell adhesion and migration were strongly suppressed. US2 was necessary and sufficient for degradation of the majority of its substrates, but remarkably, the HCMV NK cell evasion function UL141 requisitioned US2 to enhance downregulation of the NK cell ligand CD112. UL141 retained CD112 in the ER from where US2 promoted its TRC8-dependent retrotranslocation and degradation. These findings redefine US2 as a multifunctional degradation hub which, through recruitment of the cellular E3 ligase TRC8, modulates diverse immune pathways involved in antigen presentation, NK cell activation, migration and coagulation; and highlight US2’s impact on HCMV pathogenesis. As the largest human herpesvirus, HCMV is a paradigm of viral immune evasion and has evolved multiple mechanisms to evade immune detection and enable survival. The HCMV genes US2, US3, US6 and US11 promote virus persistence by their ability to downregulate cell surface MHC. We developed ‘Plasma Membrane Profiling’ (PMP), an unbiased SILAC-based proteomics technique to ask whether MHC molecules are the only focus of these genes, or whether additional cellular immunoreceptors are also targeted. PMP compares the relative abundance of cell surface receptors between control and viral gene expressing cells. We found that whereas US3, US6 and US11 were remarkably MHC specific, US2 modulated expression of a wide variety of cell surface immunoreceptors. US2-mediated proteasomal degradation of integrin α-chains blocked integrin signaling and suppressed cell adhesion and migration. All US2 substrates were degraded via the cellular E3 ligase TRC8, and in a remarkable example of cooperativity between HCMV immune-evasins, UL141 requisitioned US2 to target the NK cell ligand CD112 for proteasomal degradation. HCMV US2 and UL141 are therefore modulators of multiple immune-related pathways and act as a multifunctional degradation hub that inhibits the migration, immune recognition and killing of HCMV-infected cells.
Collapse
Affiliation(s)
- Jye-Lin Hsu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | - Peter Tomasec
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | - Eva Ruckova
- Regional Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Daniel Sugrue
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Gavin S. Wilkie
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - Andrew J. Davison
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | | | - Paul J. Lehner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
95
|
Mirjačić Martinović K, Babović N, Džodić R, Jurišić V, Matković S, Konjević G. Favorable in vitro effects of combined IL-12 and IL-18 treatment on NK cell cytotoxicity and CD25 receptor expression in metastatic melanoma patients. J Transl Med 2015; 13:120. [PMID: 25889680 PMCID: PMC4421987 DOI: 10.1186/s12967-015-0479-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/30/2015] [Indexed: 11/26/2022] Open
Abstract
Background As IL-12 and IL-18 have important immunostimulatory role the aim of this study was to investigate their in vitro effects on functional and receptor characteristics of NK cells and their subsets in healthy controls (HC) and metastatic melanoma patients (MM). Methods Peripheral blood mononuclear cells (PBMC) of HC and MM were stimulated with culture medium alone, medium supplemented with IL-12 (10 ng/ml), IL-18 (100 ng/ml) and their combination. NK cell activity was determined using radioactive cytotoxicity assay, while perforin, CD107a and pSTAT-4 expression, IFN-γ production and the expression of NKG2D, DNAM-1, CD161, CD158a/b, CD25, IL-12R beta 1/2 receptors on CD3−CD56+ NK cells and their CD3−CD56dim+ and CD3−CD56bright+ subsets were analyzed by flow cytometry. Cytokine induced level of DAP10 in PBMC was analyzed by reverse transcription polymerase chain reaction. Results IL-12 alone or in combination with IL-18 significantly induced NK cell activity and CD107a degranulation marker expression in MM and HC, while IL-18 alone did not have any effect in patients. The combination of IL-12 and IL-18 significantly increased mean fluorescence intensity (MFI) of IFN-γ in all NK cell subsets in HC and only in the bright subset in MM. MM that belong to M1c group with metastasis in liver and increased LDH serum values had significantly lower increase in NK cell cytotoxicity after combined IL-12 and IL-18 treatment compared to the patients in M1a and M1b categories. These results could be explained by decreased IL-12R expression and lower increase in pSTAT-4 and perforin expression in NK cells of M1c patients after IL-12 and combined IL-12 and IL-18 treatment. IL-18 alone significantly decreased NKG2D receptor expression and level of DAP10 signaling molecule in MM, while combined IL-12 and IL-18 increased the expression of CD25 on all NK cell subsets in HC and MM. Additionally, MM that belong to M1a + M1b group had significantly higher increase in CD25 receptor expression compared to the patients in M1c group. Conclusions The novel data obtained in this study support the use of IL-12 and IL-18 in combination for developing new therapeutic strategies for metastatic melanoma especially for patients with better survival rate and prognosis.
Collapse
Affiliation(s)
- Katarina Mirjačić Martinović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia.
| | - Nada Babović
- Department of Medical Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia.
| | - Radan Džodić
- Surgical Oncology Clinic, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia. .,School of Medicine, University of Belgrade, Dr Subotića 8, 11000, Beograd, Serbia.
| | - Vladimir Jurišić
- Faculty of Medical Sciences, University of Kragujevac, P.BOX 124, 34000, Kragujevac, Serbia.
| | - Suzana Matković
- Department of Medical Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia.
| | - Gordana Konjević
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia. .,School of Medicine, University of Belgrade, Dr Subotića 8, 11000, Beograd, Serbia.
| |
Collapse
|
96
|
Martinet L, Ferrari De Andrade L, Guillerey C, Lee JS, Liu J, Souza-Fonseca-Guimaraes F, Hutchinson DS, Kolesnik TB, Nicholson SE, Huntington ND, Smyth MJ. DNAM-1 expression marks an alternative program of NK cell maturation. Cell Rep 2015; 11:85-97. [PMID: 25818301 DOI: 10.1016/j.celrep.2015.03.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/31/2014] [Accepted: 02/27/2015] [Indexed: 12/27/2022] Open
Abstract
Natural killer (NK) cells comprise a heterogeneous population of cells important for pathogen defense and cancer surveillance. However, the functional significance of this diversity is not fully understood. Here, we demonstrate through transcriptional profiling and functional studies that the activating receptor DNAM-1 (CD226) identifies two distinct NK cell functional subsets: DNAM-1(+) and DNAM-1(-) NK cells. DNAM-1(+) NK cells produce high levels of inflammatory cytokines, have enhanced interleukin 15 signaling, and proliferate vigorously. By contrast, DNAM-1(-) NK cells that differentiate from DNAM-1(+) NK cells have greater expression of NK-cell-receptor-related genes and are higher producers of MIP1 chemokines. Collectively, our data reveal the existence of a functional program of NK cell maturation marked by DNAM-1 expression.
Collapse
Affiliation(s)
- Ludovic Martinet
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse 31000, France
| | - Lucas Ferrari De Andrade
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; Células Inflamatórias e Neoplásicas group, Universidade Federal do Paraná, Curitiba, Paraná 81530-001, Brazil
| | - Camille Guillerey
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Jason S Lee
- Control of Gene Expression Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Jing Liu
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | | | - Dana S Hutchinson
- Department of Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical, Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Tatiana B Kolesnik
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3050, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3050, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3050, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Medicine, University of Queensland, Herston, QLD 4006, Australia.
| |
Collapse
|
97
|
Martinet L, Smyth MJ. Balancing natural killer cell activation through paired receptors. Nat Rev Immunol 2015; 15:243-54. [PMID: 25743219 DOI: 10.1038/nri3799] [Citation(s) in RCA: 365] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural killer (NK) cells are innate lymphocytes that are crucial for the control of infections and malignancies. NK cells express a variety of inhibitory and activating receptors that facilitate fine discrimination between damaged and healthy cells. Among them, a family of molecules that bind nectin and nectin-like proteins has recently emerged and has been shown to function as an important regulator of NK cell functions. These molecules include CD226, T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), CD96, and cytotoxic and regulatory T cell molecule (CRTAM). In this Review, we focus on the recent advances in our understanding of how these receptors regulate NK cell biology and of their roles in pathologies such as cancer, infection and autoimmunity.
Collapse
Affiliation(s)
- Ludovic Martinet
- 1] Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia. [2] Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1037, Cancer Research Center of Toulouse, Toulouse F-31000, France
| | - Mark J Smyth
- 1] Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia. [2] School of Medicine, University of Queensland, Herston, Queensland 4006, Australia
| |
Collapse
|
98
|
|
99
|
Ribeiro ST, Ribot JC, Silva-Santos B. Five Layers of Receptor Signaling in γδ T-Cell Differentiation and Activation. Front Immunol 2015; 6:15. [PMID: 25674089 PMCID: PMC4306313 DOI: 10.3389/fimmu.2015.00015] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
The contributions of γδ T-cells to immunity to infection or tumors critically depend on their activation and differentiation into effectors capable of secreting cytokines and killing infected or transformed cells. These processes are molecularly controlled by surface receptors that capture key extracellular cues and convey downstream intracellular signals that regulate γδ T-cell physiology. The understanding of how environmental signals are integrated by γδ T-cells is critical for their manipulation in clinical settings. Here, we discuss how different classes of surface receptors impact on human and murine γδ T-cell differentiation, activation, and expansion. In particular, we review the role of five receptor types: the T-cell receptor (TCR), costimulatory receptors, cytokine receptors, NK receptors, and inhibitory receptors. Some of the key players are the costimulatory receptors CD27 and CD28, which differentially impact on pro-inflammatory subsets of γδ T-cells; the cytokine receptors IL-2R, IL-7R, and IL-15R, which drive functional differentiation and expansion of γδ T-cells; the NK receptor NKG2D and its contribution to γδ T-cell cytotoxicity; and the inhibitory receptors PD-1 and BTLA that control γδ T-cell homeostasis. We discuss these and other receptors in the context of a five-step model of receptor signaling in γδ T-cell differentiation and activation, and discuss its implications for the manipulation of γδ T-cells in immunotherapy.
Collapse
Affiliation(s)
- Sérgio T Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa , Lisboa , Portugal
| | - Julie C Ribot
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa , Lisboa , Portugal
| | - Bruno Silva-Santos
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa , Lisboa , Portugal
| |
Collapse
|
100
|
Tarazona R, Campos C, Pera A, Sanchez-Correa B, Solana R. Flow Cytometry Analysis of NK Cell Phenotype and Function in Aging. Methods Mol Biol 2015; 1343:9-18. [PMID: 26420705 DOI: 10.1007/978-1-4939-2963-4_2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Natural killer (NK) cells represent a subpopulation of lymphocytes involved in innate immunity, defined recently as group 1 of innate lymphoid cells (ILCs). NK cells are cytotoxic lymphocytes with a relevant role in the destruction of transformed cells as virus-infected or tumor cells, as well as the regulation of the immune response through cytokine and chemokine production that activates other cellular components of innate and adaptive immunity. In humans, NK cell subsets have been defined according to the level of expression of CD56. Aging differentially affects NK cell subsets and NK cell function. Here, we describe protocols for the delineation of NK cell subsets and the analysis of their functional capacity using multiparametric flow cytometry.
Collapse
Affiliation(s)
- Raquel Tarazona
- Immunology Unit, Department of Physiology, University of Extremadura, Caceres, Spain
| | - Carmen Campos
- Department of Immunology, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14071, Spain
| | - Alejandra Pera
- Department of Immunology, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14071, Spain
| | | | - Rafael Solana
- Department of Immunology, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14071, Spain.
| |
Collapse
|