1
|
Marin-Rodero M, Cintado E, Walker AJ, Jayewickreme T, Pinho-Ribeiro FA, Richardson Q, Jackson R, Chiu IM, Benoist C, Stevens B, Trejo JL, Mathis D. The meninges host a distinct compartment of regulatory T cells that preserves brain homeostasis. Sci Immunol 2025; 10:eadu2910. [PMID: 39873623 PMCID: PMC11924117 DOI: 10.1126/sciimmunol.adu2910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025]
Abstract
Our understanding of the meningeal immune system has recently burgeoned, particularly regarding how innate and adaptive effector cells are mobilized to meet brain challenges. However, information on how meningeal immunocytes guard brain homeostasis in healthy individuals remains limited. This study highlights the heterogeneous, polyfunctional regulatory T cell (Treg) compartment in the meninges. A Treg subtype specialized in controlling interferon-γ (IFN-γ) responses and another dedicated to regulating follicular B cell responses were substantial components of this compartment. Accordingly, punctual Treg ablation rapidly unleashed IFN-γ production by meningeal lymphocytes, unlocked access to the brain parenchyma, and altered meningeal B cell profiles. Distally, the hippocampus assumed a reactive state, with morphological and transcriptional changes in multiple glial cell types. Within the dentate gyrus, neural stem cells underwent more death and were blocked from further differentiation, which coincided with impairments in short-term spatial-reference memory. Thus, meningeal Tregs are a multifaceted safeguard of brain homeostasis at steady state.
Collapse
Affiliation(s)
| | - Elisa Cintado
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas; Madrid, Spain
| | - Alec J. Walker
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | | | | | | | - Ruaidhrí Jackson
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| | - Isaac M. Chiu
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| | | | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital; Boston, MA, USA
| | - José Luís Trejo
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas; Madrid, Spain
| | - Diane Mathis
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| |
Collapse
|
2
|
Lee KY, Mei Y, Liu H, Schwarz H. CD137-expressing regulatory T cells in cancer and autoimmune diseases. Mol Ther 2025; 33:51-70. [PMID: 39668561 PMCID: PMC11764688 DOI: 10.1016/j.ymthe.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining immune homeostasis, with critical roles in preventing aberrant immune responses that occur in autoimmune diseases and chronic inflammation. Conversely, the abundance of Tregs in cancer is associated with impaired anti-tumor immunity, and tumor immune evasion. Recent work demonstrates that CD137, a well-known costimulatory molecule for T cells, is highly expressed on Tregs in pathological conditions, while its expression is minimal or negligible on peripheral Tregs. The expression of CD137 marks Tregs with potent immunosuppressive phenotype that foster cancer progression and are protective against certain autoimmune diseases. Hence CD137 has emerged as a marker for Tregs. However, several important questions still remain regarding the expression and function of CD137 in Tregs. Here, we provide an overview of our current knowledge of Treg mechanisms of action, with a focus on the role of CD137 in modulating Treg activity. We also explore the implications of CD137+ Tregs in both cancer and autoimmune diseases, emphasizing the significance of targeting these cells for therapeutic intervention in these conditions.
Collapse
Affiliation(s)
- Kang Yi Lee
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Yu Mei
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Haiyan Liu
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore.
| | - Herbert Schwarz
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
| |
Collapse
|
3
|
Duléry R, Piccinelli S, Beg MS, Jang JE, Romee R. Haploidentical hematopoietic cell transplantation as a platform for natural killer cell immunotherapy. Am J Hematol 2024; 99:2340-2350. [PMID: 39248561 DOI: 10.1002/ajh.27471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024]
Abstract
An innovative approach is crucially needed to manage relapse after allogeneic hematopoietic cell transplantation (HCT) in patients with advanced hematological malignancies. This review explores key aspects of haploidentical HCT with post-transplant cyclophosphamide, highlighting the potential and suitability of this platform for natural killer (NK) cell immunotherapy. NK cells, known for their unique abilities to eliminate cancer cells, can also exhibit memory-like features and enhanced cytotoxicity when activated by cytokines. By discussing promising results from clinical trials, the review delves into the recent major advances: donor-derived NK cells can be expanded ex vivo in large numbers, cytokine activation may enhance NK cell persistence and efficacy in vivo, and post-HCT NK cell infusion can improve outcomes in high-risk and/or relapsed myeloid malignancies without increasing the risk of graft-versus-host disease, severe cytokine release syndrome, or neurotoxicity. Looking ahead, cytokine-activated NK cells can be synergized with immunomodulatory agents and/or genetically engineered to enhance their tumor-targeting specificity, cytotoxicity, and persistence while preventing exhaustion. The ongoing exploration of these strategies holds promising preliminary results and could be rapidly translated into clinical applications for the benefit of the patients.
Collapse
Affiliation(s)
- Rémy Duléry
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Piccinelli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Ji Eun Jang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Rizwan Romee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Wang N, Wang G, Feng X, Yang T. Identification of Immune Gene Signature Associated with T Cells and Natural Killer Cells in Type 1 Diabetes. Diabetes Metab Syndr Obes 2024; 17:2983-2996. [PMID: 39139741 PMCID: PMC11321349 DOI: 10.2147/dmso.s470738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
Purpose This study aimed to investigate the abnormal infiltration of immune cells in type 1 diabetes mellitus (T1D) and elucidate their regulatory mechanisms. Methods Public T1D-related gene expression data were obtained from the Gene Expression Omnibus database.The GSE123658 dataset analyzed whole blood RNA-seq data from type 1 diabetic patients and healthy volunteers. The GSE110914 dataset analyzed neutrophils purified from peripheral blood of patients with symptomatic and pre-symptomatic type 1 diabetes (T1D), at risk of T1D, and healthy controls. Immune cell infiltration analysis was performed to identify abnormally infiltrating immune cells. Differentially expressed immune genes (DEIGs) in T1D samples were identified, followed by the construction of an immune gene signature (IGS) using a protein-protein interaction (PPI) network and Least absolute shrinkage and selection operator Cox regression analyses (LASSO Cox regression analyses). The regulatory mechanisms underlying IGS were explored using gene set enrichment analysis. Furthermore, expression validation, diagnostic efficacy evaluation, and upstream miRNA prediction of hub signature genes were performed. We verified the miRNA expression of the key gene colony stimulating factor 1 (CSF1) and microRNA-326 (miR-326) by reverse transcription-quantitative PCR (RT‒qPCR). Results The proportion of infiltrating T and natural killer (NK) cells differed between the T1D and control samples, and 207 immune genes (IGs) related to these immune cells were extracted. After differential expression, PPI, and LASSO Cox regression analyses, four signature DEIGs were identified for IGS construction: notch receptor 1 (NOTCH1), Janus kinase 3 (JAK3), tumor necrosis factor receptor superfamily member 4(TNFRSF4), and CSF1. Key pathways such as the Toll-like receptor signaling pathway were significantly activated in the high-risk group. Moreover, the upregulation of CSF1 in T1D samples was confirmed using a validation dataset, and CSF1 showed high diagnostic efficacy for T1D. Furthermore, CSF1 was targeted by miR-326.We used validated key genes in T1D patients, several of which were confirmed by RT‒qPCR. Conclusion In conclusion, the identified key IGs may play an important role in T1D. CSF1 can be developed as a novel diagnostic biomarker for T1D.
Collapse
Affiliation(s)
- Na Wang
- Department of Endocrinology, Lianyungang Clinical College of Nanjing Medical University, Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Guofeng Wang
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Xiuli Feng
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Teng Yang
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| |
Collapse
|
5
|
Marin-Rodero M, Reyes EC, Walker AJ, Jayewickreme T, Pinho-Ribeiro FA, Richardson Q, Jackson R, Chiu IM, Benoist C, Stevens B, Trejo JL, Mathis D. The meninges host a unique compartment of regulatory T cells that bulwarks adult hippocampal neurogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599387. [PMID: 38948783 PMCID: PMC11212874 DOI: 10.1101/2024.06.17.599387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Our knowledge about the meningeal immune system has recently burgeoned, particularly our understanding of how innate and adaptive effector cells are mobilized to meet brain challenges. However, information on how meningeal immunocytes guard brain homeostasis in healthy individuals remains sparse. This study highlights the heterogeneous and polyfunctional regulatory-T (Treg) cell compartment in the meninges. A Treg subtype specialized in controlling Th1-cell responses and another known to control responses in B-cell follicles were substantial components of this compartment, foretelling that punctual Treg-cell ablation rapidly unleashed interferon-gamma production by meningeal lymphocytes, unlocked their access to the brain parenchyma, and altered meningeal B-cell profiles. Distally, the hippocampus assumed a reactive state, with morphological and transcriptional changes in multiple glial-cell types; within the dentate gyrus, neural stem cells showed exacerbated death and desisted from further differentiation, associated with inhibition of spatial-reference memory. Thus, meningeal Treg cells are a multifaceted bulwark to brain homeostasis at steady-state. One sentence summary A distinct population of regulatory T cells in the murine meninges safeguards homeostasis by keeping local interferon-γ-producing lymphocytes in check, thereby preventing their invasion of the parenchyma, activation of hippocampal glial cells, death of neural stem cells, and memory decay.
Collapse
|
6
|
Jou E. Clinical and basic science aspects of innate lymphoid cells as novel immunotherapeutic targets in cancer treatment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 209:1-60. [PMID: 39461748 DOI: 10.1016/bs.pmbts.2024.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Immunotherapy has revolutionised cancer treatment over the past decade, demonstrating remarkable efficacy across a broad range of cancer types. However, not all patients or cancer types respond to contemporary clinically-utilised immunotherapeutic strategies, which largely focus on harnessing adaptive immune T cells for cancer treatment. Accordingly, it is increasingly recognised that upstream innate immune pathways, which govern and orchestrate the downstream adaptive immune response, may prove critical in overcoming cancer immunotherapeutic resistance. Innate lymphoid cells (ILCs) are the most recently discovered major innate immune cell population. They have overarching roles in homeostasis and orchestrating protective immunity against pathogens. As innate immune counterparts of adaptive immune T cells, ILCs exert effector functions through the secretion of cytokines and direct cell-to-cell contact, with broad influence on the overall immune response. Importantly, dysregulation of ILC subsets have been associated with a range of diseases, including immunodeficiency disorders, allergy, autoimmunity, and more recently, cancer. ILCs may either promote or inhibit cancer initiation and progression depending on the cancer type and the specific ILC subsets involved. Critically, therapeutic targeting of ILCs and their associated cytokines shows promise against a wide range of cancer types in both preclinical models and early phase oncology clinical trials. This chapter provides a comprehensive overview of the current understanding of ILC subsets and the associated cytokines they produce in cancer pathogenesis, with specific focus on how these innate pathways are, or can be targeted, therapeutically to overcome therapeutic resistance and ultimately improve patient care.
Collapse
Affiliation(s)
- Eric Jou
- Department of Oncology, Oxford University Hospitals, University of Oxford, Oxford, United Kingdom; Kellogg College, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
7
|
Nersesian S, Carter EB, Lee SN, Westhaver LP, Boudreau JE. Killer instincts: natural killer cells as multifactorial cancer immunotherapy. Front Immunol 2023; 14:1269614. [PMID: 38090565 PMCID: PMC10715270 DOI: 10.3389/fimmu.2023.1269614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Natural killer (NK) cells integrate heterogeneous signals for activation and inhibition using germline-encoded receptors. These receptors are stochastically co-expressed, and their concurrent engagement and signaling can adjust the sensitivity of individual cells to putative targets. Against cancers, which mutate and evolve under therapeutic and immunologic pressure, the diversity for recognition provided by NK cells may be key to comprehensive cancer control. NK cells are already being trialled as adoptive cell therapy and targets for immunotherapeutic agents. However, strategies to leverage their naturally occurring diversity and agility have not yet been developed. In this review, we discuss the receptors and signaling pathways through which signals for activation or inhibition are generated in NK cells, focusing on their roles in cancer and potential as targets for immunotherapies. Finally, we consider the impacts of receptor co-expression and the potential to engage multiple pathways of NK cell reactivity to maximize the scope and strength of antitumor activities.
Collapse
Affiliation(s)
- Sarah Nersesian
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Emily B. Carter
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Stacey N. Lee
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | - Jeanette E. Boudreau
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
8
|
Langston PK, Sun Y, Ryback BA, Mueller AL, Spiegelman BM, Benoist C, Mathis D. Regulatory T cells shield muscle mitochondria from interferon-γ-mediated damage to promote the beneficial effects of exercise. Sci Immunol 2023; 8:eadi5377. [PMID: 37922340 PMCID: PMC10860652 DOI: 10.1126/sciimmunol.adi5377] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/02/2023] [Indexed: 11/05/2023]
Abstract
Exercise enhances physical performance and reduces the risk of many disorders such as cardiovascular disease, type 2 diabetes, dementia, and cancer. Exercise characteristically incites an inflammatory response, notably in skeletal muscles. Although some effector mechanisms have been identified, regulatory elements activated in response to exercise remain obscure. Here, we have addressed the roles of Foxp3+CD4+ regulatory T cells (Tregs) in the healthful activities of exercise via immunologic, transcriptomic, histologic, metabolic, and biochemical analyses of acute and chronic exercise models in mice. Exercise rapidly induced expansion of the muscle Treg compartment, thereby guarding against overexuberant production of interferon-γ and consequent metabolic disruptions, particularly mitochondrial aberrancies. The performance-enhancing effects of exercise training were dampened in the absence of Tregs. Thus, exercise is a natural Treg booster with therapeutic potential in disease and aging contexts.
Collapse
Affiliation(s)
- P. Kent Langston
- Department of Immunology, Harvard Medical School; Boston, 02115, USA
| | - Yizhi Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute; Boston, 02115, USA
- Department of Cell Biology, Harvard Medical School; Boston, 02115, USA
| | - Birgitta A. Ryback
- Department of Cancer Biology, Dana-Farber Cancer Institute; Boston, 02115, USA
| | - Amber L. Mueller
- Department of Genetics, Harvard Medical School; Boston, 02115, USA
| | - Bruce M. Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute; Boston, 02115, USA
- Department of Cell Biology, Harvard Medical School; Boston, 02115, USA
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School; Boston, 02115, USA
| |
Collapse
|
9
|
Ou Q, Power R, Griffin MD. Revisiting regulatory T cells as modulators of innate immune response and inflammatory diseases. Front Immunol 2023; 14:1287465. [PMID: 37928540 PMCID: PMC10623442 DOI: 10.3389/fimmu.2023.1287465] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Regulatory T cells (Treg) are known to be critical for the maintenance of immune homeostasis by suppressing the activation of auto- or allo-reactive effector T cells through a diverse repertoire of molecular mechanisms. Accordingly, therapeutic strategies aimed at enhancing Treg numbers or potency in the setting of autoimmunity and allogeneic transplants have been energetically pursued and are beginning to yield some encouraging outcomes in early phase clinical trials. Less well recognized from a translational perspective, however, has been the mounting body of evidence that Treg directly modulate most aspects of innate immune response under a range of different acute and chronic disease conditions. Recognizing this aspect of Treg immune modulatory function provides a bridge for the application of Treg-based therapies to common medical conditions in which organ and tissue damage is mediated primarily by inflammation involving myeloid cells (mononuclear phagocytes, granulocytes) and innate lymphocytes (NK cells, NKT cells, γδ T cells and ILCs). In this review, we comprehensively summarize pre-clinical and human research that has revealed diverse modulatory effects of Treg and specific Treg subpopulations on the range of innate immune cell types. In each case, we emphasize the key mechanistic insights and the evidence that Treg interactions with innate immune effectors can have significant impacts on disease severity or treatment. Finally, we discuss the opportunities and challenges that exist for the application of Treg-based therapeutic interventions to three globally impactful, inflammatory conditions: type 2 diabetes and its end-organ complications, ischemia reperfusion injury and atherosclerosis.
Collapse
Affiliation(s)
- Qifeng Ou
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Rachael Power
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
- Nephrology Department, Galway University Hospitals, Saolta University Healthcare Group, Galway, Ireland
| |
Collapse
|
10
|
Lee M, Bell CJM, Rubio Garcia A, Godfrey L, Pekalski M, Wicker LS, Todd JA, Ferreira RC. CD56 bright natural killer cells preferentially kill proliferating CD4 + T cells. DISCOVERY IMMUNOLOGY 2023; 2:kyad012. [PMID: 37649552 PMCID: PMC10465185 DOI: 10.1093/discim/kyad012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/14/2023] [Accepted: 08/10/2023] [Indexed: 09/01/2023]
Abstract
Human CD56br natural killer (NK) cells represent a small subset of CD56+ NK cells in circulation and are largely tissue-resident. The frequency and number of CD56br NK cells in blood has been shown to increase following administration of low-dose IL-2 (LD-IL2), a therapy aimed to specifically expand CD4+ regulatory T cells (Tregs). Given the potential clinical application of LD-IL-2 immunotherapy across several immune diseases, including the autoimmune disease type 1 diabetes, a better understanding of the functional consequences of this expansion is urgently needed. In this study, we developed an in vitro co-culture assay with activated CD4+ T cells to measure NK cell killing efficiency. We show that CD56br and CD56dim NK cells show similar efficiency at killing activated CD4+ conventional T (Tconv) and Treg cell subsets. However, in contrast to CD56dim cells, CD56br NK cells preferentially target highly proliferative cells. We hypothesize that CD56br NK cells have an immunoregulatory role through the elimination of proliferating autoreactive CD4+ Tconv cells that have escaped Treg suppression. These results have implications for the interpretation of current and future trials of LD-IL-2 by providing evidence for a new, possibly beneficial immunomodulatory mechanism of LD-IL-2-expanded CD56br NK cells.
Collapse
Affiliation(s)
- Mercede Lee
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Charles J M Bell
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Arcadio Rubio Garcia
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Leila Godfrey
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Marcin Pekalski
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Linda S Wicker
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - John A Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Ricardo C Ferreira
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| |
Collapse
|
11
|
Möller KJ, Wegner LHM, Malsy J, Baumdick ME, Borggrewe M, Jordan-Paiz A, Jung JM, Martrus G, Kretschmer P, Sagebiel AF, Schreurs RRCE, Hagen SH, Burmester G, Clauditz TS, Pals ST, Boettcher M, Melling N, Sauter G, Tomuschat C, Königs I, Schumacher U, Altfeld M, Bernink JH, Perez D, Reinshagen K, Bunders MJ. Expanded ILC2s in human infant intestines promote tissue growth. Mucosal Immunol 2023; 16:408-421. [PMID: 37121384 DOI: 10.1016/j.mucimm.2023.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/02/2023]
Abstract
Early life is characterized by extraordinary challenges, including rapid tissue growth and immune adaptation to foreign antigens after birth. During this developmental stage, infants have an increased risk of immune-mediated diseases. Here, we demonstrate that tissue-resident, interleukin (IL)-13- and IL-4-producing group 2 innate lymphoid cells (ILC2s) are enriched in human infant intestines compared to adult intestines. Organoid systems were employed to assess the role of infant intestinal ILC2s in intestinal development and showed that IL-13 and IL-4 increased epithelial cell proliferation and skewed cell differentiation toward secretory cells. IL-13 furthermore upregulated the production of mediators of type-2 immunity by infant intestinal epithelial cells, including vascular endothelial growth factor-A and IL-26, a chemoattractant for eosinophils. In line with these in vitro findings increased numbers of eosinophils were detected in vivo in infant intestines. Taken together, ILC2s are enriched in infant intestines and can support intestinal development while inducing an epithelial secretory response associated with type 2 immune-mediated diseases.
Collapse
Affiliation(s)
- Kimberly J Möller
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Lucy H M Wegner
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Jakob Malsy
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany; I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany
| | - Martin E Baumdick
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Malte Borggrewe
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ana Jordan-Paiz
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Johannes M Jung
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Glòria Martrus
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Paul Kretschmer
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Adrian F Sagebiel
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany; Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renée R C E Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Sven H Hagen
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Gunter Burmester
- Department of Pediatric Gastroenterology, Altonaer Children's Hospital, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altonaer Children's Hospital, Hamburg, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Tomuschat
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altonaer Children's Hospital, Hamburg, Germany
| | - Ingo Königs
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altonaer Children's Hospital, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Altfeld
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Jochem H Bernink
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Asklepios General Hospital-Altona, Hamburg, Germany
| | - Konard Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altonaer Children's Hospital, Hamburg, Germany
| | - Madeleine J Bunders
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
12
|
Abstract
INTRODUCTION New methods in cancer immunotherapy, such as chimeric antigen receptor (CAR)-T cells, have shown promising results in destroying malignant cells. However, limitations and side effects of CAR-T cell therapy, such as graft-versus-host disease (GVHD), neurotoxicity, and cytokine release syndrome, have motivated researchers to investigate safer alternative cells like natural killer (NK) cells. AREA COVERED NK cells can effectively recognize hematologic malignant cells and destroy them. Many clinical and preclinical studies investigate the efficacy of CAR-NK cells in treating lymphoma and other hematologic malignancies. The results of published clinical trials and preclinical studies have shown that CAR-NK cells could be an appropriate choice for treating lymphoma. In this review, we discuss the characteristics of CAR-NK cells, their role in treating B-cell and T-cell lymphoma, and the challenges faced by using them. We also highlight clinical trials using CAR-NK cells for treating lymphoma. EXPERT OPINION CAR-NK cells have shown promising results in cancer therapy, especially B-cell lymphoma, with a much lower risk for GVHD, cytokine release syndrome, and neurotoxicity than CAR-T cells. Further investigations are required to overcome the obstacles of CAR-NK cell therapy, both generally, and in cancers like T-cell lymphoma.
Collapse
Affiliation(s)
- Shaghayegh Khanmohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Chen Z, Tong L, Neo SY, Li S, Gao J, Schlisio S, Lundqvist A. CD25 bright NK cells display superior function and metabolic activity under regulatory T cell-mediated suppression. Oncoimmunology 2023; 12:2175517. [PMID: 36970070 PMCID: PMC10038043 DOI: 10.1080/2162402x.2023.2175517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Infusion of natural killer (NK) cells is an attractive therapeutic modality in patients with cancer. However, the activity of NK cells is regulated by several mechanisms operating within solid tumors. Regulatory T (Treg) cells suppress NK cell activity through various mechanisms including deprivation of IL-2 via the IL-2 receptor alpha (CD25). Here, we investigate CD25 expression on NK cells to confer persistence in Treg cells containing solid tumor models of renal cell carcinoma (RCC). Compared with IL-2, stimulation with IL-15 increases the expression of CD25 resulting in enhanced response to IL-2 as evidenced by increased phosphorylation of STAT5. Compared with CD25dim NK cells, CD25bright NK cells isolated from IL-15 primed NK cells display increased proliferative and metabolic activity as well as increased ability to persist in Treg cells containing RCC tumor spheroids. These results support strategies to enrich for or selectively expand CD25bright NK cells for adoptive cellular therapy of NK cells.
Collapse
Affiliation(s)
- Ziqing Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, New Jersey, USA
| | - Le Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Shi Yong Neo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Singapore Immunology Network, Agency for Science, Technology and Research, Republic of Singapore
| | - Shuijie Li
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jiwei Gao
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Schlisio
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- CONTACT Andreas Lundqvist Department of Oncology-Pathology, Karolinska Institutet, Solna17164, Sweden
| |
Collapse
|
14
|
Seliger B, Koehl U. Underlying mechanisms of evasion from NK cells as rational for improvement of NK cell-based immunotherapies. Front Immunol 2022; 13:910595. [PMID: 36045670 PMCID: PMC9422402 DOI: 10.3389/fimmu.2022.910595] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Natural killer (NK) cells belong to the family of innate immune cells with the capacity to recognize and kill tumor cells. Different phenotypes and functional properties of NK cells have been described in tumor patients, which could be shaped by the tumor microenvironment. The discovery of HLA class I-specific inhibitory receptors controlling NK cell activity paved the way to the fundamental concept of modulating immune responses that are regulated by an array of inhibitory receptors, and emphasized the importance to explore the potential of NK cells in cancer therapy. Although a whole range of NK cell-based approaches are currently being developed, there are still major challenges that need to be overcome for improved efficacy of these therapies. These include escape of tumor cells from NK cell recognition due to their expression of inhibitory molecules, immune suppressive signals of NK cells, reduced NK cell infiltration of tumors, an immune suppressive micromilieu and limited in vivo persistence of NK cells. Therefore, this review provides an overview about the NK cell biology, alterations of NK cell activities, changes in tumor cells and the tumor microenvironment contributing to immune escape or immune surveillance by NK cells and their underlying molecular mechanisms as well as the current status and novel aspects of NK cell-based therapeutic strategies including their genetic engineering and their combination with conventional treatment options to overcome tumor-mediated evasion strategies and improve therapy efficacy.
Collapse
Affiliation(s)
- Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- *Correspondence: Barbara Seliger,
| | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
- Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| |
Collapse
|
15
|
Regulatory T cells in skeletal muscle repair and regeneration: recent insights. Cell Death Dis 2022; 13:680. [PMID: 35931697 PMCID: PMC9356005 DOI: 10.1038/s41419-022-05142-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 01/21/2023]
Abstract
Skeletal muscle repair and regeneration after injury is a multi-stage process, involving a dynamic inflammatory microenvironment consisting of a complex network formed by the interaction of immune cells and their secreted cytokines. The homeostasis of the inflammatory microenvironment determines whether skeletal muscle repair tissues will ultimately form scar tissue or regenerative tissue. Regulatory T cells (Tregs) regulate homeostasis within the immune system and self-immune tolerance, and play a crucial role in skeletal muscle repair and regeneration. Dysregulated Tregs function leads to abnormal repair. In this review, we discuss the role and mechanisms of Tregs in skeletal muscle repair and regeneration after injury and provide new strategies for Treg immunotherapy in skeletal muscle diseases.
Collapse
|
16
|
Ruzgys P, Navickaitė D, Palepšienė R, Uždavinytė D, Barauskaitė N, Novickij V, Girkontaitė I, Šitkauskienė B, Šatkauskas S. Induction of Bystander and Abscopal Effects after Electroporation-Based Treatments. Cancers (Basel) 2022; 14:3770. [PMID: 35954434 PMCID: PMC9367330 DOI: 10.3390/cancers14153770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/21/2022] [Accepted: 07/30/2022] [Indexed: 02/01/2023] Open
Abstract
Electroporation-based antitumor therapies, including bleomycin electrotransfer, calcium electroporation, and irreversible electroporation, are very effective on directly treated tumors, but have no or low effect on distal nodules. In this study, we aimed to investigate the abscopal effect following calcium electroporation and bleomycin electrotransfer and to find out the effect of the increase of IL-2 serum concentration by muscle transfection. The bystander effect was analyzed in in vitro studies on 4T1tumor cells, while abscopal effect was investigated in an in vivo setting using Balb/c mice bearing 4T1 tumors. ELISA was used to monitor IL-2 serum concentration. We showed that, similarly to cell treatment with bleomycin electrotransfer, the bystander effect occurs also following calcium electroporation and that these effects can be combined. Combination of these treatments also resulted in the enhancement of the abscopal effect in vivo. Since these treatments resulted in an increase of IL-2 serum concentration only in mice bearing one but not two tumors, we increased IL-2 serum concentration by muscle transfection. Although this did not enhance the abscopal effect of combined tumor treatment using calcium electroporation and bleomycin electrotransfer, boosting of IL-2 serum concentration had a significant inhibitory effect on directly treated tumors.
Collapse
Affiliation(s)
- Paulius Ruzgys
- Biophysical Research Group, Vytautas Magnus University, Vileikos St. 8, LT-44404 Kaunas, Lithuania; (P.R.); (D.N.); (R.P.); (D.U.); (N.B.)
| | - Diana Navickaitė
- Biophysical Research Group, Vytautas Magnus University, Vileikos St. 8, LT-44404 Kaunas, Lithuania; (P.R.); (D.N.); (R.P.); (D.U.); (N.B.)
| | - Rūta Palepšienė
- Biophysical Research Group, Vytautas Magnus University, Vileikos St. 8, LT-44404 Kaunas, Lithuania; (P.R.); (D.N.); (R.P.); (D.U.); (N.B.)
| | - Dovilė Uždavinytė
- Biophysical Research Group, Vytautas Magnus University, Vileikos St. 8, LT-44404 Kaunas, Lithuania; (P.R.); (D.N.); (R.P.); (D.U.); (N.B.)
| | - Neringa Barauskaitė
- Biophysical Research Group, Vytautas Magnus University, Vileikos St. 8, LT-44404 Kaunas, Lithuania; (P.R.); (D.N.); (R.P.); (D.U.); (N.B.)
| | - Vitalij Novickij
- Faculty of Electronics, Vilnius Gediminas Technical University, Saulėtekio al. 11, LT-10223 Vilnius, Lithuania;
| | - Irutė Girkontaitė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania;
| | - Brigita Šitkauskienė
- Department of Immunology and Allergology, Medical Academy, Lithuanian University of Health Sciences, Eiveniu 2, LT-50161 Kaunas, Lithuania;
| | - Saulius Šatkauskas
- Biophysical Research Group, Vytautas Magnus University, Vileikos St. 8, LT-44404 Kaunas, Lithuania; (P.R.); (D.N.); (R.P.); (D.U.); (N.B.)
| |
Collapse
|
17
|
Bai R, Cui J. Burgeoning Exploration of the Role of Natural Killer Cells in Anti-PD-1/PD-L1 Therapy. Front Immunol 2022; 13:886931. [PMID: 35634343 PMCID: PMC9133458 DOI: 10.3389/fimmu.2022.886931] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Antibodies targeting programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) have been considered breakthrough therapies for a variety of solid and hematological malignancies. Although cytotoxic T cells play an important antitumor role during checkpoint blockade, they still show a potential killing effect on tumor types showing loss of/low major histocompatibility complex (MHC) expression and/or low neoantigen load; this knowledge has shifted the focus of researchers toward mechanisms of action other than T cell-driven immune responses. Evidence suggests that the blockade of the PD-1/PD-L1 axis may also improve natural killer (NK)-cell function and activity through direct or indirect mechanisms, which enhances antitumor cytotoxic effects; although important, this topic has been neglected in previous studies. Recently, some studies have reported evidence of PD-1 and PD-L1 expression in human NK cells, performed exploration of the intrinsic mechanism by which PD-1/PD-L1 blockade enhances NK-cell responses, and made some progress. This article summarizes the recent advances regarding the expression of PD-1 and PD-L1 molecules on the surface of NK cells as well as the interaction between anti-PD-1/PD-L1 drugs and NK cells and associated molecular mechanisms in the tumor microenvironment.
Collapse
Affiliation(s)
| | - Jiuwei Cui
- *Correspondence: Jiuwei Cui, ; orcid.org/0000-0001-6496-7550
| |
Collapse
|
18
|
Tarannum M, Romee R, Shapiro RM. Innovative Strategies to Improve the Clinical Application of NK Cell-Based Immunotherapy. Front Immunol 2022; 13:859177. [PMID: 35401529 PMCID: PMC8990319 DOI: 10.3389/fimmu.2022.859177] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022] Open
Abstract
Natural killer cells constitute a part of the innate immune system that mediates an effective immune response towards virus-infected and malignant cells. In recent years, research has focused on exploring and advancing NK cells as an active immunotherapy platform. Despite major advances, there are several key challenges that need to be addressed for the effective translation of NK cell research to clinical applications. This review highlights some of these challenges and the innovative strategies being developed to overcome them, including in vitro expansion, in vivo persistence, infiltration to the tumor site, and prevention of exhaustion.
Collapse
Affiliation(s)
- Mubin Tarannum
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Rizwan Romee
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Roman M Shapiro
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
19
|
Muralidhara P, Sood V, Vinayak Ashok V, Bansal K. Pregnancy and Tumour: The Parallels and Differences in Regulatory T Cells. Front Immunol 2022; 13:866937. [PMID: 35493450 PMCID: PMC9043683 DOI: 10.3389/fimmu.2022.866937] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/15/2022] [Indexed: 11/21/2022] Open
Abstract
Immunological tolerance plays a critical role during pregnancy as semi-allogeneic fetus must be protected from immune responses during the gestational period. Regulatory T cells (Tregs), a subpopulation of CD4+ T cells that express transcription factor Foxp3, are central to the maintenance of immunological tolerance and prevention of autoimmunity. Tregs are also known to accumulate at placenta in uterus during pregnancy, and they confer immunological tolerance at maternal-fetal interface by controlling the immune responses against alloantigens. Thus, uterine Tregs help in maintaining an environment conducive for survival of the fetus during gestation, and low frequency or dysfunction of Tregs is associated with recurrent spontaneous abortions and other pregnancy-related complications such as preeclampsia. Interestingly, there are many parallels in the development of placenta and solid tumours, and the tumour microenvironment is considered to be somewhat similar to that at maternal-fetal interface. Moreover, Tregs play a largely similar role in tumour immunity as they do at placenta- they create a tolerogenic system and suppress the immune responses against the cells within tumour and at maternal-fetal interface. In this review, we discuss the role of Tregs in supporting the proper growth of the embryo during pregnancy. We also highlight the similarities and differences between Tregs at maternal-fetal interface and tumour Tregs, in an attempt to draw a comparison between their roles in these two physiologic and pathologic states.
Collapse
Affiliation(s)
| | | | | | - Kushagra Bansal
- Molecular Biology and Genetics Unit (MBGU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
| |
Collapse
|
20
|
Gold nanorods enhance different immune cells and allow for efficient targeting of CD4+ Foxp3+ Tregulatory cells. PLoS One 2021; 16:e0241882. [PMID: 34460818 PMCID: PMC8404976 DOI: 10.1371/journal.pone.0241882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 07/27/2021] [Indexed: 01/22/2023] Open
Abstract
Gold nanoparticles (AuNPs) hold great promise in nanomedicine, yet their successful clinical translation has not been realized. Some challenges include effective AuNP targeting and delivery to improve modulation of immune cells of interest while limiting potential adverse effects. In order to overcome these challenges, we must fully understand how AuNPs impact different immune cell subsets, particularly within the dendritic cell and T cell compartments. Herein, we show that polyethylene glycol coated (PEG) gold nanorods (AuNRs) and PEG AuNRs covered with a thin layer of silver (AuNR/Ag) may enhance the immune response towards immune suppression or activation. We also studied the ability to enhance CD4+ Foxp3+ Tregs in vitro using AuNRs functionalized with interleukin 2 (IL2), a cytokine that is important in Treg development and homeostasis. Our results indicate that AuNRs enhance different immune cells and that NP composition matters in immune targeting. This knowledge will help us understand how to better design AuNRs to target and enhance the immune system.
Collapse
|
21
|
Host genetic control of natural killer cell diversity revealed in the Collaborative Cross. Proc Natl Acad Sci U S A 2021; 118:2018834118. [PMID: 33649222 DOI: 10.1073/pnas.2018834118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Natural killer (NK) cells are innate effectors armed with cytotoxic and cytokine-secreting capacities whose spontaneous antitumor activity is key to numerous immunotherapeutic strategies. However, current mouse models fail to mirror the extensive immune system variation that exists in the human population which may impact on NK cell-based therapies. We performed a comprehensive profiling of NK cells in the Collaborative Cross (CC), a collection of novel recombinant inbred mouse strains whose genetic diversity matches that of humans, thereby providing a unique and highly diverse small animal model for the study of immune variation. We demonstrate that NK cells from CC strains displayed a breadth of phenotypic and functional variation reminiscent of that reported for humans with regards to cell numbers, key marker expression, and functional capacities. We took advantage of the vast genetic diversity of the CC and identified nine genomic loci through quantitative trait locus mapping driving these phenotypic variations. SNP haplotype patterns and variant effect analyses identified candidate genes associated with lung NK cell numbers, frequencies of CD94+ NK cells, and expression levels of NKp46. Thus, we demonstrate that the CC represents an outstanding resource to study NK cell diversity and its regulation by host genetics.
Collapse
|
22
|
Single-cell analysis of FOXP3 deficiencies in humans and mice unmasks intrinsic and extrinsic CD4 + T cell perturbations. Nat Immunol 2021; 22:607-619. [PMID: 33833438 PMCID: PMC8173714 DOI: 10.1038/s41590-021-00910-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/26/2021] [Indexed: 01/31/2023]
Abstract
FOXP3 deficiency in mice and in patients with immune dysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome results in fatal autoimmunity by altering regulatory T (Treg) cells. CD4+ T cells in patients with IPEX syndrome and Foxp3-deficient mice were analyzed by single-cell cytometry and RNA-sequencing, revealing heterogeneous Treg-like cells, some very similar to normal Treg cells, others more distant. Conventional T cells showed no widespread activation or helper T cell bias, but a monomorphic disease signature affected all CD4+ T cells. This signature proved to be cell extrinsic since it was extinguished in mixed bone marrow chimeric mice and heterozygous mothers of patients with IPEX syndrome. Normal Treg cells exerted dominant suppression, quenching the disease signature and revealing in mutant Treg-like cells a small cluster of genes regulated cell-intrinsically by FOXP3, including key homeostatic regulators. We propose a two-step pathogenesis model: cell-intrinsic downregulation of core FOXP3-dependent genes destabilizes Treg cells, de-repressing systemic mediators that imprint the disease signature on all T cells, furthering Treg cell dysfunction. Accordingly, interleukin-2 treatment improved the Treg-like compartment and survival.
Collapse
|
23
|
Shaver KA, Croom-Perez TJ, Copik AJ. Natural Killer Cells: The Linchpin for Successful Cancer Immunotherapy. Front Immunol 2021; 12:679117. [PMID: 33995422 PMCID: PMC8115550 DOI: 10.3389/fimmu.2021.679117] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/09/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy is a highly successful and rapidly evolving treatment modality that works by augmenting the body’s own immune system. While various immune stimulation strategies such as PD-1/PD-L1 or CTLA-4 checkpoint blockade result in robust responses, even in patients with advanced cancers, the overall response rate is low. While immune checkpoint inhibitors are known to enhance cytotoxic T cells’ antitumor response, current evidence suggests that immune responses independent of cytotoxic T cells, such as Natural Killer (NK) cells, play crucial role in the efficacy of immunotherapeutic interventions. NK cells hold a distinct role in potentiating the innate immune response and activating the adaptive immune system. This review highlights the importance of the early actions of the NK cell response and the pivotal role NK cells hold in priming the immune system and setting the stage for successful response to cancer immunotherapy. Yet, in many patients the NK cell compartment is compromised thus lowering the chances of successful outcomes of many immunotherapies. An overview of mechanisms that can drive NK cell dysfunction and hinder immunotherapy success is provided. Rather than relying on the likely dysfunctional endogenous NK cells to work with immunotherapies, adoptive allogeneic NK cell therapies provide a viable solution to increase response to immunotherapies. This review highlights the advances made in development of NK cell therapeutics for clinical application with evidence supporting their combinatorial application with other immune-oncology approaches to improve outcomes of immunotherapies.
Collapse
Affiliation(s)
- Kari A Shaver
- College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Tayler J Croom-Perez
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Alicja J Copik
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
24
|
Low-dose interleukin-2-loaded nanoparticle effect on NK and T-reg cell expression in experimentally induced type 1 diabetes mellitus. GASTROENTEROLOGY REVIEW 2021; 16:67-82. [PMID: 33986891 PMCID: PMC8112267 DOI: 10.5114/pg.2021.104737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
Introduction Type 1 diabetes mellitus is an autoimmune disorder characterized by inflammatory damage to pancreatic β cells resulting in loss of insulin secretion. In autoimmune type 1 diabetes mellitus (T1D) natural killer cells (NK) initiate pancreatic islets cell lyses in autoimmune T1D. Loss of T regulatory cells (Treg) at disease onset facilitates the activation and accumulation of NKs in the pancreatic microenvironment. A proper low-dose interleukin 2 (IL-2) could enhance Tregs and enforce control and regulation of pro-inflammatory NKs. Aim This relation needs to be studied to improve therapeutic strategies aimed at resetting the balance between Tregs and proinflammatory cells. Material and methods We used novel formulations of low-dose IL-2 loaded on chitosan nanoparticles. The study included 116 T1D BALB/c mice experimentally induced by streptozotocin, divided into groups. Their splenocytes were maintained in a short-term culture for assessment of expression of CD4+FOXP3+ Treg and NKp46+NK by both flow cytometry and enzyme-linked immunoassay (ELISA). Morphological, immunohistochemical, and morphometrical analyses were done.In vitro suppressor assay was used to assess the suppressor effect of Treg cells after exogenous IL-2 treatment. Results NK cell expression, NKp46 level, and NK cell functions were modulated more in mice injected with IL-2-loaded chitosan nanoparticles than in other groups. A statistical inverse correlation was found between Treg and NK cell expression in IL-2-loaded chitosan with 0.3 µIU (p = 0.047), and this correlation was related to FOXP3 expression on Treg cells. The modified expression of NK and NKp46 was noticed in mice injected with 0.3 µIU for longer duration (3 weeks) (p < 0.001), but the NK functions did not show any significant changes with prolonged treatment. Conclusions Prolonged administration of low-dose IL-2 results in the vigorous expression of NKp46, indicating a significant role of Tregs in NK stimulation and motivation. Low-dose IL-2 selectively modulates NKp46 NK and FOXP3+ Tregs and increases their expression.
Collapse
|
25
|
Kumar V. Innate Lymphoid Cells and Adaptive Immune Cells Cross-Talk: A Secret Talk Revealed in Immune Homeostasis and Different Inflammatory Conditions. Int Rev Immunol 2021; 40:217-251. [PMID: 33733998 DOI: 10.1080/08830185.2021.1895145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The inflammatory immune response has evolved to protect the host from different pathogens, allergens, and endogenous death or damage-associated molecular patterns. Both innate and adaptive immune components are crucial in inducing an inflammatory immune response depending on the stimulus type and its duration of exposure or the activation of the primary innate immune response. As the source of inflammation is removed, the aggravated immune response comes to its homeostatic level. However, the failure of the inflammatory immune response to subside to its normal level generates chronic inflammatory conditions, including autoimmune diseases and cancer. Innate lymphoid cells (ILCs) are newly discovered innate immune cells, which are present in abundance at mucosal surfaces, including lungs, gastrointestinal tract, and reproductive tract. Also, they are present in peripheral blood circulation, skin, and lymph nodes. They play a crucial role in generating the pro-inflammatory immune response during diverse conditions. On the other hand, adaptive immune cells, including different types of T and B cells are major players in the pathogenesis of autoimmune diseases (type 1 diabetes mellitus, rheumatoid arthritis, psoriasis, and systemic lupus erythematosus, etc.) and cancers. Thus the article is designed to discuss the immunological role of different ILCs and their interaction with adaptive immune cells in maintaining the immune homeostasis, and during inflammatory autoimmune diseases along with other inflammatory conditions (excluding pathogen-induced inflammation), including cancer, graft-versus-host diseases, and human pregnancy.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, St Lucia, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
26
|
Lopes JE, Fisher JL, Flick HL, Wang C, Sun L, Ernstoff MS, Alvarez JC, Losey HC. ALKS 4230: a novel engineered IL-2 fusion protein with an improved cellular selectivity profile for cancer immunotherapy. J Immunother Cancer 2021; 8:jitc-2020-000673. [PMID: 32317293 PMCID: PMC7204809 DOI: 10.1136/jitc-2020-000673] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Interleukin-2 (IL-2) plays a pivotal role in immune homeostasis due to its ability to stimulate numerous lymphocyte subsets including natural killer (NK) cells, effector CD4+ and CD8+ T cells, and regulatory T cells (Tregs). Low concentrations of IL-2 induce signaling through the high-affinity IL-2 receptor (IL-2R) comprised of IL-2Rα, IL-2Rβ, and common γ chain (γc), preferentially expressed on Tregs. Higher concentrations of IL-2 are necessary to induce signaling through the intermediate-affinity IL-2R, composed of IL-2Rβ and γc, expressed on memory CD8+ T cells and NK cells. Recombinant human IL-2 (rhIL-2) is approved for treatment of metastatic melanoma and renal cell carcinoma (RCC), but adverse events including capillary leak syndrome, potentially mediated through interaction with the high-affinity IL-2R, limit its therapeutic use. Furthermore, antitumor efficacy of IL-2 may also be limited by preferential expansion of immunosuppressive Tregs. ALKS 4230 is an engineered fusion protein comprised of a circularly-permuted IL-2 with the extracellular domain of IL-2Rα, designed to selectively activate effector lymphocytes bearing the intermediate-affinity IL-2R. RESULTS ALKS 4230 was equipotent to rhIL-2 in activating human cells bearing the intermediate-affinity IL-2R, and less potent than rhIL-2 on cells bearing the high-affinity IL-2R. As observed in vitro with primary human cells from healthy donors and advanced cancer patients, ALKS 4230 induced greater activation and expansion of NK cells with reduced expansion of Tregs relative to rhIL-2. Similarly, in mice, ALKS 4230 treatment stimulated greater expansion of NK cells and memory-phenotype CD8+ T cells at doses that did not expand or activate Tregs. ALKS 4230 treatment induced significantly lower levels of proinflammatory cytokines, including tumor necrosis factor alpha, interleukin-6, and interferon gamma relative to rhIL-2. Furthermore, ALKS 4230 exhibited superior antitumor efficacy in the mouse B16F10 lung tumor model, where ALKS 4230 could be administered via multiple routes of administration and dosing schedules while achieving equivalent antitumor efficacy. CONCLUSIONS ALKS 4230 exhibited enhanced pharmacokinetic and selective pharmacodynamic properties resulting in both improved antitumor efficacy and lower indices of toxicity relative to rhIL-2 in mice. These data highlight the potential of ALKS 4230 as a novel cancer immunotherapy, and as such, the molecule is being evaluated clinically.
Collapse
Affiliation(s)
- Jared E Lopes
- Research, Alkermes, Inc, Waltham, Massachusetts, USA
| | - Jan L Fisher
- Department of Medicine, The Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, New Hampshire, USA
| | | | - Chunhua Wang
- Research, Alkermes, Inc, Waltham, Massachusetts, USA
| | - Lei Sun
- Research, Alkermes, Inc, Waltham, Massachusetts, USA
| | - Marc S Ernstoff
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Juan C Alvarez
- Computational and Structural Chemistry, Merck & Co, Boston, Massachusetts, USA
| | | |
Collapse
|
27
|
Kim H, Perovanovic J, Shakya A, Shen Z, German CN, Ibarra A, Jafek JL, Lin NP, Evavold BD, Chou DHC, Jensen PE, He X, Tantin D. Targeting transcriptional coregulator OCA-B/Pou2af1 blocks activated autoreactive T cells in the pancreas and type 1 diabetes. J Exp Med 2021; 218:e20200533. [PMID: 33295943 PMCID: PMC7731945 DOI: 10.1084/jem.20200533] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/27/2020] [Accepted: 10/09/2020] [Indexed: 11/04/2022] Open
Abstract
The transcriptional coregulator OCA-B promotes expression of T cell target genes in cases of repeated antigen exposure, a necessary feature of autoimmunity. We hypothesized that T cell-specific OCA-B deletion and pharmacologic OCA-B inhibition would protect mice from autoimmune diabetes. We developed an Ocab conditional allele and backcrossed it onto a diabetes-prone NOD/ShiLtJ strain background. T cell-specific OCA-B loss protected mice from spontaneous disease. Protection was associated with large reductions in islet CD8+ T cell receptor specificities associated with diabetes pathogenesis. CD4+ clones associated with diabetes were present but associated with anergic phenotypes. The protective effect of OCA-B loss was recapitulated using autoantigen-specific NY8.3 mice but diminished in monoclonal models specific to artificial or neoantigens. Rationally designed membrane-penetrating OCA-B peptide inhibitors normalized glucose levels and reduced T cell infiltration and proinflammatory cytokine expression in newly diabetic NOD mice. Together, the results indicate that OCA-B is a potent autoimmune regulator and a promising target for pharmacologic inhibition.
Collapse
MESH Headings
- Alleles
- Amino Acid Sequence
- Animals
- Autoantigens/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Crosses, Genetic
- Cytokines/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/prevention & control
- Disease Models, Animal
- Female
- Gene Deletion
- Germ Cells/metabolism
- Humans
- Inflammation Mediators/metabolism
- Lymph Nodes/metabolism
- Lymphocyte Activation
- Male
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Ovalbumin
- Pancreas/metabolism
- Pancreas/pathology
- Peptides/pharmacology
- Receptors, Antigen, T-Cell/metabolism
- Spleen/pathology
- T-Lymphocytes/immunology
- Trans-Activators/deficiency
- Trans-Activators/metabolism
- Transcription, Genetic
- Mice
Collapse
Affiliation(s)
- Heejoo Kim
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Jelena Perovanovic
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Arvind Shakya
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Zuolian Shen
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Cody N German
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Andrea Ibarra
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Jillian L Jafek
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Nai-Pin Lin
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - Brian D Evavold
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Danny H-C Chou
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - Peter E Jensen
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Xiao He
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Dean Tantin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
28
|
Effect of low dose IL-2 loaded chitosan nanoparticles on natural killer and regulatory T cell expression in experimentally induced autoimmune type 1 diabetes mellitus. Cent Eur J Immunol 2021; 45:382-392. [PMID: 33658887 PMCID: PMC7882410 DOI: 10.5114/ceji.2020.103412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 12/13/2019] [Indexed: 11/20/2022] Open
Abstract
Introduction Natural killer cells (NK) initiate pancreatic islets cell lyses in autoimmune type 1 diabetes mellitus (T1D). Loss of T regulatory cells (Treg) at disease onset facilitates activation and accumulation of NKs in the pancreatic microenvironment. A proper low dose interleukin 2 (IL-2) could enhance Tregs and enforce control and regulation of pro-inflammatory NKs. This relation needs to be studied to improve therapeutic strategies aimed at resetting the balance between Tregs and proinflammatory cells. Material and methods We used novel formulations of low dose IL-2 loaded on chitosan nanoparticles. The study included 116 T1D BALB/c mice experimentally induced by streptozotocin, divided into groups. Their splenocytes were maintained in a short-term culture for assessment of expression of CD4+Foxp3+ Treg and NKp46+NK by both flow cytometry and enzyme linked immunoassay (ELISA). In vitro suppressor-assay was used in order to assess the suppressor effect of Treg cells after exogenous IL-2 treatment. Results NK cell expression, NKp46 level and NK cell functions were modulated in mice injected with IL-2 loaded chitosan nanoparticles than other groups. A statistical inverse correlation was found between Treg and NK cell expression in IL-2 loaded chitosan with (0.3 µIU) (p = 0.047) and this correlation was related to Foxp3 expression on Treg cells. The modified expression of NK and NKp46 was noticed in mice injected with (0.3 µIU) for longer duration (three weeks) (p < 0.001) but the NK functions did not show any significant changes with prolonged treatment. Conclusions Low dose (0.3) µIU IL-2 nanoparticles effectively modulated NK and NKp46 expression. It selectively modulates the suppressive activity of Tregs indicating a significant role of Tregs in NK activation and function by controlling the availability of IL-2 in the microenvironment.
Collapse
|
29
|
Stephen ZR, Zhang M. Recent Progress in the Synergistic Combination of Nanoparticle-Mediated Hyperthermia and Immunotherapy for Treatment of Cancer. Adv Healthc Mater 2021; 10:e2001415. [PMID: 33236511 PMCID: PMC8034553 DOI: 10.1002/adhm.202001415] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has demonstrated great clinical success in certain cancers, driven primarily by immune checkpoint blockade and adoptive cell therapies. Immunotherapy can elicit strong, durable responses in some patients, but others do not respond, and to date immunotherapy has demonstrated success in only a limited number of cancers. To address this limitation, combinatorial approaches with chemo- and radiotherapy have been applied in the clinic. Extensive preclinical evidence suggests that hyperthermia therapy (HT) has considerable potential to augment immunotherapy with minimal toxicity. This progress report will provide a brief overview of immunotherapy and HT approaches and highlight recent progress in the application of nanoparticle (NP)-based HT in combination with immunotherapy. NPs allow for tumor-specific targeting of deep tissue tumors while potentially providing more even heating. NP-based HT increases tumor immunogenicity and tumor permeability, which improves immune cell infiltration and creates an environment more responsive to immunotherapy, particularly in solid tumors.
Collapse
Affiliation(s)
- Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, Department of Neurological Surgery, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
30
|
Increased Serum Interleukin-2 Levels Are Associated with Abnormal Peripheral Blood Natural Killer Cell Levels in Patients with Active Rheumatoid Arthritis. Mediators Inflamm 2020; 2020:6108342. [PMID: 33013198 PMCID: PMC7512106 DOI: 10.1155/2020/6108342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/30/2020] [Accepted: 08/24/2020] [Indexed: 12/29/2022] Open
Abstract
Objective To investigate the relationship between serum interleukin-2 (IL-2) levels and disease activity, absolute numbers of peripheral lymphocyte subsets, autoantibodies, and associated cytokines in patients with rheumatoid arthritis (RA). Methods This study included 106 patients with RA, evaluated their disease activity (DAS28 score), and divided them into disease remission (DAS28 ≤ 2.6), low disease activity (DAS28 ≤ 3.2), and moderate-high disease activity (DAS28 > 3.2) groups. Flow cytometry was used to detect the absolute numbers of peripheral lymphocyte subpopulations and CD4+ T cell subsets in each group, and serum cytokine levels were measured using cytometric bead array. Results Serum IL-2 levels in RA patients were positively correlated with disease activity and rheumatoid factor titers (p < 0.001 and p = 0.045, respectively), and multiple regression analysis revealed that serum IL-2 levels were an independent factor affecting disease activity. Serum IL-2 levels were positively correlated with Th17/Treg ratios (p = 0.013). Compared with the remission group, peripheral lymphocyte and CD4+ T lymphocyte subsets in patients with active RA decreased to varying degrees; however, the numbers of peripheral natural killer (NK) cells were significantly higher in the moderate-high disease activity group than in the remission (p = 0.046) and low disease activity (p = 0.020) groups; the percentages of NK cells had the same trend. In addition, the number and percentage of NK cells were positively correlated with serum IL-2 levels (p = 0.018 and p = 0.006, respectively). Conclusions In RA patients, serum IL-2 levels were not only correlated with patients' disease activity and autoantibody levels but were also involved in their Th17/Treg immune imbalance. In addition, in patients with active RA, NK cell levels were abnormally elevated, possibly due to high serum levels of IL-2.
Collapse
|
31
|
Khoryati L, Pham MN, Sherve M, Kumari S, Cook K, Pearson J, Bogdani M, Campbell DJ, Gavin MA. An IL-2 mutein engineered to promote expansion of regulatory T cells arrests ongoing autoimmunity in mice. Sci Immunol 2020; 5:eaba5264. [PMID: 32817295 PMCID: PMC7643170 DOI: 10.1126/sciimmunol.aba5264] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/17/2020] [Indexed: 12/18/2022]
Abstract
Interleukin-2 (IL-2) controls the homeostasis and function of regulatory T (Treg) cells, and defects in the IL-2 pathway contribute to multiple autoimmune diseases. Although recombinant IL-2 therapy has been efficacious in certain inflammatory conditions, the capacity for IL-2 to also activate inflammatory effector responses highlights the need for IL-2-based therapeutics with improved Treg cell specificity. From a panel of rationally designed murine IL-2 variants, we identified IL-2 muteins with reduced potency and enhanced Treg cell selectivity due to increased dependence on the IL-2 receptor component CD25. As an Fc-fused homodimer, the optimal Fc.IL-2 mutein induced selective Treg cell enrichment and reduced agonism of effector cells across a wide dose range. Furthermore, despite being a weaker agonist, overall Treg cell growth was greater and more sustained due to reduced receptor-mediated clearance of the Fc.IL-2 mutein compared with Fc-fused wild-type IL-2. Preferential Treg cell enrichment was also observed in the presence of activated pathogenic T cells in the pancreas of nonobese diabetic (NOD) mice, despite a loss of Treg cell selectivity in an IL-2R proximal response. These properties facilitated potent and extended resolution of NOD diabetes with infrequent dosing schedules.
Collapse
Affiliation(s)
- Liliane Khoryati
- Immunology Research Program, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Minh Nguyet Pham
- Immunology Research Program, Benaroya Research Institute, Seattle, WA 98101, USA
| | - McKenna Sherve
- Immunology Research Program, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Swarnima Kumari
- Immunology Research Program, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Kevin Cook
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA 94080, USA
| | - Josh Pearson
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA 94080, USA
| | - Marika Bogdani
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Daniel J Campbell
- Immunology Research Program, Benaroya Research Institute, Seattle, WA 98101, USA.
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Marc A Gavin
- Immunology Research Program, Benaroya Research Institute, Seattle, WA 98101, USA.
| |
Collapse
|
32
|
Fung TH, Yang KY, Lui KO. An emerging role of regulatory T-cells in cardiovascular repair and regeneration. Theranostics 2020; 10:8924-8938. [PMID: 32802172 PMCID: PMC7415793 DOI: 10.7150/thno.47118] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence has demonstrated that immune cells play an important role in the regulation of tissue repair and regeneration. After injury, danger signals released by the damaged tissue trigger the initial pro-inflammatory phase essential for removing pathogens or cellular debris that is later replaced by the anti-inflammatory phase responsible for tissue healing. On the other hand, impaired immune regulation can lead to excessive scarring and fibrosis that could be detrimental for the restoration of organ function. Regulatory T-cells (Treg) have been revealed as the master regulator of the immune system that have both the immune and regenerative functions. In this review, we will summarize their immune role in the induction and maintenance of self-tolerance; as well as their regenerative role in directing tissue specific response for repair and regeneration. The latter is clearly demonstrated when Treg enhance the differentiation of stem or progenitor cells such as satellite cells to replace the damaged skeletal muscle, as well as the proliferation of parenchymal cells including neonatal cardiomyocytes for functional regeneration. Moreover, we will also discuss the reparative and regenerative role of Treg with a particular focus on blood vessels and cardiac tissues. Last but not least, we will describe the ongoing clinical trials with Treg in the treatment of autoimmune diseases that could give clinically relevant insights into the development of Treg therapy targeting tissue repair and regeneration.
Collapse
|
33
|
Chauhan SKS, Koehl U, Kloess S. Harnessing NK Cell Checkpoint-Modulating Immunotherapies. Cancers (Basel) 2020; 12:E1807. [PMID: 32640575 PMCID: PMC7408278 DOI: 10.3390/cancers12071807] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
During the host immune response, the precise balance of the immune system, regulated by immune checkpoint, is required to avoid infection and cancer. These immune checkpoints are the mainstream regulator of the immune response and are crucial for self-tolerance. During the last decade, various new immune checkpoint molecules have been studied, providing an attractive path to evaluate their potential role as targets for effective therapeutic interventions. Checkpoint inhibitors have mainly been explored in T cells until now, but natural killer (NK) cells are a newly emerging target for the determination of checkpoint molecules. Simultaneously, an increasing number of therapeutic dimensions have been explored, including modulatory and inhibitory checkpoint molecules, either causing dysfunction or promoting effector functions. Furthermore, the combination of the immune checkpoint with other NK cell-based therapeutic strategies could also strengthen its efficacy as an antitumor therapy. In this review, we have undertaken a comprehensive review of the literature to date regarding underlying mechanisms of modulatory and inhibitory checkpoint molecules.
Collapse
Affiliation(s)
| | - Ulrike Koehl
- Institute of cellular therapeutics, Hannover Medical School, 30625 Hannover, Germany; (U.K.); (S.K.)
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig, 04103 Leipzig, Germany
| | - Stephan Kloess
- Institute of cellular therapeutics, Hannover Medical School, 30625 Hannover, Germany; (U.K.); (S.K.)
| |
Collapse
|
34
|
Kumar V. Innate lymphoid cell and adaptive immune cell cross-talk: A talk meant not to forget. J Leukoc Biol 2020; 108:397-417. [PMID: 32557732 DOI: 10.1002/jlb.4mir0420-500rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 12/17/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a relatively new class of innate immune cells with phenotypical characters of lymphocytes but genotypically or functionally behave as typical innate immune cells. They have been classically divided into 3 groups (group 1 ILCs or ILC1s, group 2 ILCs or ILC2s, and group 3 ILCs or ILC3s). They serve as the first line of defense against invading pathogens and allergens at mucosal surfaces. The adaptive immune response works effectively in association with innate immunity as innate immune cells serve as APCs to directly stimulate the adaptive immune cells (various sets of T and B cells). Additionally, innate immune cells also secrete various effector molecules, including cytokines or chemokines impacting the function, differentiation, proliferation, and reprogramming among adaptive immune cells to maintain immune homeostasis. Only superantigens do not require their processing by innate immune cells as they are recognized directly by T cells and B cells. Thus, a major emphasis of the current article is to describe the cross-talk between different ILCs and adaptive immune cells during different conditions varying from normal physiological situations to different infectious diseases to allergic asthma.
Collapse
Affiliation(s)
- V Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
35
|
Alhabbab RY. Targeting Cancer Stem Cells by Genetically Engineered Chimeric Antigen Receptor T Cells. Front Genet 2020; 11:312. [PMID: 32391048 PMCID: PMC7188929 DOI: 10.3389/fgene.2020.00312] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
The term cancer stem cell (CSC) starts 25 years ago with the evidence that CSC is a subpopulation of tumor cells that have renewal ability and can differentiate into several distinct linages. Therefore, CSCs play crucial role in the initiation and the maintenance of cancer. Moreover, it has been proposed throughout several studies that CSCs are behind the failure of the conventional chemo-/radiotherapy as well as cancer recurrence due to their ability to resist the therapy and their ability to re-regenerate. Thus, the need for targeted therapy to eliminate CSCs is crucial; for that reason, chimeric antigen receptor (CAR) T cells has currently been in use with high rate of success in leukemia and, to some degree, in patients with solid tumors. This review outlines the most common CSC populations and their common markers, in particular CD133, CD90, EpCAM, CD44, ALDH, and EGFRVIII, the interaction between CSCs and the immune system, CAR T cell genetic engineering and signaling, CAR T cells in targeting CSCs, and the barriers in using CAR T cells as immunotherapy to treat solid cancers.
Collapse
Affiliation(s)
- Rowa Y. Alhabbab
- Division of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
36
|
Dean JW, Peters LD, Fuhrman CA, Seay HR, Posgai AL, Stimpson SE, Brusko MA, Perry DJ, Yeh WI, Newby BN, Haller MJ, Muir AB, Atkinson MA, Mathews CE, Brusko TM. Innate inflammation drives NK cell activation to impair Treg activity. J Autoimmun 2020; 108:102417. [PMID: 32035746 PMCID: PMC7086400 DOI: 10.1016/j.jaut.2020.102417] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
Abstract
IL-12 and IL-18 synergize to promote TH1 responses and have been implicated as accelerators of autoimmune pathogenesis in type 1 diabetes (T1D). We investigated the influence of these cytokines on immune cells involved in human T1D progression: natural killer (NK) cells, regulatory T cells (Tregs), and cytotoxic T lymphocytes (CTL). NK cells from T1D patients exhibited higher surface CD226 versus controls and lower CD25 compared to first-degree relatives and controls. Changes in NK cell phenotype towards terminal differentiation were associated with cytomegalovirus (CMV) seropositivity, while possession of IL18RAP, IFIH1, and IL2RA T1D-risk variants impacted NK cell activation as evaluated by immuno-expression quantitative trait loci (eQTL) analyses. IL-12 and IL-18 stimulated NK cells from healthy donors exhibited enhanced specific killing of myelogenous K562 target cells. Moreover, activated NK cells increased expression of NKG2A, NKG2D, CD226, TIGIT and CD25, which enabled competition for IL-2 upon co-culture with Tregs, resulting in Treg downregulation of FOXP3, production of IFNγ, and loss of suppressive function. We generated islet-autoreactive CTL "avatars", which upon exposure to IL-12 and IL-18, upregulated IFNγ and Granzyme-B leading to increased lymphocytotoxicity of a human β-cell line in vitro. These results support a model for T1D pathogenesis wherein IL-12 and IL-18 synergistically enhance CTL and NK cell cytotoxic activity and disrupt immunoregulation by Tregs.
Collapse
MESH Headings
- Adolescent
- Adult
- Biomarkers
- Cells, Cultured
- Child
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Diabetes Mellitus, Type 1/etiology
- Diabetes Mellitus, Type 1/metabolism
- Disease Susceptibility
- Female
- Humans
- Immunity, Innate
- Immunophenotyping
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Male
- Middle Aged
- Models, Biological
- Phenotype
- Quantitative Trait Loci
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Young Adult
Collapse
Affiliation(s)
- Joseph W Dean
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA; Department of Infectious Disease and Immunology, University of Florida, Gainesville, FL, USA
| | - Leeana D Peters
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Christopher A Fuhrman
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA; NanoString Technologies, Seattle, WA, USA
| | - Howard R Seay
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA; BD Biosciences, Ashland, OR, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Scott E Stimpson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Maigan A Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Wen-I Yeh
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA; BD Biosciences, Ashland, OR, USA
| | - Brittney N Newby
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA; Fate Therapeutics, San Diego, CA, USA
| | - Michael J Haller
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA; Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew B Muir
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Mark A Atkinson
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Clayton E Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA; Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA; Department of Pediatrics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
37
|
Sakaguchi S, Mikami N, Wing JB, Tanaka A, Ichiyama K, Ohkura N. Regulatory T Cells and Human Disease. Annu Rev Immunol 2020; 38:541-566. [PMID: 32017635 DOI: 10.1146/annurev-immunol-042718-041717] [Citation(s) in RCA: 654] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Naturally occurring CD4+ regulatory T cells (Tregs), which specifically express the transcription factor FoxP3 in the nucleus and CD25 and CTLA-4 on the cell surface, are a functionally distinct T cell subpopulation actively engaged in the maintenance of immunological self-tolerance and homeostasis. Recent studies have facilitated our understanding of the cellular and molecular basis of their generation, function, phenotypic and functional stability, and adaptability. It is under investigation in humans how functional or numerical Treg anomalies, whether genetically determined or environmentally induced, contribute to immunological diseases such as autoimmune diseases. Also being addressed is how Tregs can be targeted to control physiological and pathological immune responses, for example, by depleting them to enhance tumor immunity or by expanding them to treat immunological diseases. This review discusses our current understanding of Treg immunobiology in normal and disease states, with a perspective on the realization of Treg-targeting therapies in the clinic.
Collapse
Affiliation(s)
- Shimon Sakaguchi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; .,Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Norihisa Mikami
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - James B Wing
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - Atsushi Tanaka
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - Kenji Ichiyama
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - Naganari Ohkura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan;
| |
Collapse
|
38
|
Mancusi A, Piccinelli S, Velardi A, Pierini A. CD4 +FOXP3 + Regulatory T Cell Therapies in HLA Haploidentical Hematopoietic Transplantation. Front Immunol 2019; 10:2901. [PMID: 31921162 PMCID: PMC6927932 DOI: 10.3389/fimmu.2019.02901] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/26/2019] [Indexed: 12/11/2022] Open
Abstract
Since their discovery CD4+FOXP3+ regulatory T cells (Tregs) represented a promising tool to induce tolerance in allogeneic hematopoietic cell transplantation. Preclinical models proved that adoptive transfer of Tregs or the use of compounds that can favor their function in vivo are effective for prevention and treatment of graft-vs.-host disease (GvHD). Following these findings, Treg-based therapies have been employed in clinical trials. Adoptive immunotherapy with Tregs effectively prevents GvHD induced by alloreactive T cells in the setting of one HLA haplotype mismatched hematopoietic transplantation. The absence of post transplant pharmacologic immunosuppression unleashes T-cell mediated graft-vs.-tumor (GvT) effect, which results in an unprecedented, almost complete control of leukemia relapse in this setting. In the present review, we will report preclinical studies and clinical trials that demonstrate Treg ability to promote donor engraftment, protect from GvHD and improve GvT effect. We will also discuss new strategies to further enhance in vivo efficacy of Treg-based therapies.
Collapse
Affiliation(s)
- Antonella Mancusi
- Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, Perugia, Italy
| | - Sara Piccinelli
- Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, Perugia, Italy
| | - Andrea Velardi
- Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, Perugia, Italy
| | - Antonio Pierini
- Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
39
|
Sivori S, Meazza R, Quintarelli C, Carlomagno S, Della Chiesa M, Falco M, Moretta L, Locatelli F, Pende D. NK Cell-Based Immunotherapy for Hematological Malignancies. J Clin Med 2019; 8:E1702. [PMID: 31623224 PMCID: PMC6832127 DOI: 10.3390/jcm8101702] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/31/2022] Open
Abstract
Natural killer (NK) lymphocytes are an integral component of the innate immune system and represent important effector cells in cancer immunotherapy, particularly in the control of hematological malignancies. Refined knowledge of NK cellular and molecular biology has fueled the interest in NK cell-based antitumor therapies, and recent efforts have been made to exploit the high potential of these cells in clinical practice. Infusion of high numbers of mature NK cells through the novel graft manipulation based on the selective depletion of T cells and CD19+ B cells has resulted into an improved outcome in children with acute leukemia given human leucocyte antigen (HLA)-haploidentical hematopoietic transplantation. Likewise, adoptive transfer of purified third-party NK cells showed promising results in patients with myeloid malignancies. Strategies based on the use of cytokines or monoclonal antibodies able to induce and optimize NK cell activation, persistence, and expansion also represent a novel field of investigation with remarkable perspectives of favorably impacting on outcome of patients with hematological neoplasia. In addition, preliminary results suggest that engineering of mature NK cells through chimeric antigen receptor (CAR) constructs deserve further investigation, with the goal of obtaining an "off-the-shelf" NK cell bank that may serve many different recipients for granting an efficient antileukemia activity.
Collapse
Affiliation(s)
- Simona Sivori
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy (S.C.); (M.D.C.)
- Centre of Excellence for Biomedical Research, University of Genoa, 16132 Genoa, Italy
| | - Raffaella Meazza
- Department of Integrated Oncological Therapies, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
| | - Concetta Quintarelli
- Department of Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, 00165 Rome, Italy; (C.Q.); (F.L.)
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Simona Carlomagno
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy (S.C.); (M.D.C.)
| | - Mariella Della Chiesa
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy (S.C.); (M.D.C.)
- Centre of Excellence for Biomedical Research, University of Genoa, 16132 Genoa, Italy
| | - Michela Falco
- Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy;
| | - Franco Locatelli
- Department of Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, 00165 Rome, Italy; (C.Q.); (F.L.)
- Department of Gynecology/Obstetrics and Pediatrics, Sapienza University, 00185 Rome, Italy
| | - Daniela Pende
- Department of Integrated Oncological Therapies, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
| |
Collapse
|
40
|
Spolski R, Li P, Leonard WJ. Biology and regulation of IL-2: from molecular mechanisms to human therapy. Nat Rev Immunol 2019; 18:648-659. [PMID: 30089912 DOI: 10.1038/s41577-018-0046-y] [Citation(s) in RCA: 401] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
IL-2 was first identified as a growth factor capable of driving the expansion of activated human T cell populations. In the more than 40 years since its discovery, a tremendous amount has been learned regarding the mechanisms that regulate the expression of both IL-2 and its cell surface receptor, its mechanisms of signalling and its range of biological actions. More recently, the mechanisms by which IL-2 regulates CD4+ T cell differentiation and function have been elucidated. IL-2 also regulates the effector and memory responses of CD8+ T cells, and the loss of IL-2 or responsiveness to IL-2 at least in part explains the exhausted phenotype that occurs during chronic viral infections and in tumour responses. These basic mechanistic studies have led to the therapeutic ability to manipulate the action of IL-2 on regulatory T (Treg) cells for the treatment of autoimmune disease and on CD8+ T cells for immunotherapy of cancer. IL-2 can have either positive or deleterious effects, and we discuss here recent ideas and approaches for manipulating the actions and overall net effects of IL-2 in disease settings, including cancer.
Collapse
Affiliation(s)
- Rosanne Spolski
- Laboratory of Molecular Immunology and The Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peng Li
- Laboratory of Molecular Immunology and The Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Warren J Leonard
- Laboratory of Molecular Immunology and The Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
41
|
Functional interactions between innate lymphoid cells and adaptive immunity. Nat Rev Immunol 2019; 19:599-613. [PMID: 31350531 PMCID: PMC6982279 DOI: 10.1038/s41577-019-0194-8] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 12/19/2022]
Abstract
Innate lymphoid cells (ILCs) are enriched at barrier surfaces of the mammalian body where they rapidly respond to host, microbial or environmental stimuli to promote immunity or tissue homeostasis. Furthermore, ILCs are dysregulated in multiple human diseases. Over the past decade, substantial advances have been made in identifying the heterogeneity and functional diversity of ILCs, which have revealed striking similarities to T cell subsets. However, emerging evidence indicates that ILCs also have a complex role in directly influencing the adaptive immune response in the context of development, homeostasis, infection or inflammation. In turn, adaptive immunity reciprocally regulates ILCs, which indicates that these interactions are a crucial determinant of immune responses within tissues. Here, we summarize our current understanding of functional interactions between ILCs and the adaptive immune system, discuss limitations and future areas of investigation, and consider the potential for these interactions to be therapeutically harnessed to benefit human health.
Collapse
|
42
|
Huang Y, Mao K, Germain RN. Thinking differently about ILCs-Not just tissue resident and not just the same as CD4 + T-cell effectors. Immunol Rev 2019; 286:160-171. [PMID: 30294968 DOI: 10.1111/imr.12704] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/10/2018] [Indexed: 12/16/2022]
Abstract
Innate lymphoid cells (ILCs) resemble adaptive T lymphocytes based on transcription factor expression, cytokine production, and their presumptive roles in immunity, but are activated for effector function through cytokine signaling and not antigen-specific receptors. The prevailing view is that ILCs adapt to specific microenvironments during development and operate as tissue-resident cells in co-operation with antigen-specific T cells to provide host protection and contribute to tissue maintenance. In particular, conventional models equate the activity of different ILC subsets with CD4+ effector T-cell types based on corresponding transcription factor expression and a potential for comparable cytokine production. Based on recent data from our laboratory, we suggest that these views on tissue residence and parallel functioning to CD4+ T cells are too restrictive. Our findings show that ILC2s can be mobilized from the gut under inflammatory conditions and contribute to distal immunity in the lungs during infection, whereas gut-resident ILC3s operate in a quite distinct manner from Th17 CD4+ effector cells in responding to commensal microbes, with important implications for control of metabolic homeostasis. In this review, we discuss the recent advances leading to these revised views of ILC inter-organ trafficking and the distinct and complementary function of ILCs with respect to adaptive T cells in establishing and maintaining a physiologic host environment.
Collapse
Affiliation(s)
- Yuefeng Huang
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National institute of Health, Bethesda, Maryland.,Department of Microbiology & Immunology, College of Physicians & Surgeons, Columbia University, New York, New York
| | - Kairui Mao
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National institute of Health, Bethesda, Maryland
| | - Ronald N Germain
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National institute of Health, Bethesda, Maryland
| |
Collapse
|
43
|
Abstract
Natural killer (NK) cells have evolved to complement T and B cells in host defense against pathogens and cancer. They recognize infected cells and tumors using a sophisticated array of activating, costimulatory, and inhibitory receptors that are expressed on NK cell subsets to create extensive functional diversity. NK cells can be targeted to kill with exquisite antigen specificity by antibody-dependent cellular cytotoxicity. NK and T cells share many of the costimulatory and inhibitory receptors that are currently under evaluation in the clinic for cancer immunotherapy. As with T cells, genetic engineering is being employed to modify NK cells to specifically target them to tumors and to enhance their effector functions. As the selective pressures exerted by immunotherapies to augment CD8+T cell responses may result in loss of MHC class I, NK cells may provide an important fail-safe to eliminate these tumors by their capacity to eliminate tumors that are “missing self.”
Collapse
Affiliation(s)
- Jeffrey S. Miller
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, California 94143, USA
- The Parker Institute for Cancer Immunotherapy, San Francisco, California 94143, USA
| |
Collapse
|
44
|
Sagebiel AF, Steinert F, Lunemann S, Körner C, Schreurs RRCE, Altfeld M, Perez D, Reinshagen K, Bunders MJ. Tissue-resident Eomes + NK cells are the major innate lymphoid cell population in human infant intestine. Nat Commun 2019; 10:975. [PMID: 30816112 PMCID: PMC6395753 DOI: 10.1038/s41467-018-08267-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/18/2018] [Indexed: 01/20/2023] Open
Abstract
Innate lymphoid cells (ILC), including natural killer (NK) cells, are implicated in host-defense and tissue-growth. However, the composition and kinetics of NK cells in the intestine during the first year of life, when infants are first broadly exposed to exogenous antigens, are still unclear. Here we show that CD103+ NK cells are the major ILC population in the small intestines of infants. When compared to adult intestinal NK cells, infant intestinal NK cells exhibit a robust effector phenotype, characterized by Eomes, perforin and granzyme B expression, and superior degranulation capacity. Absolute intestinal NK cell numbers decrease gradually during the first year of life, coinciding with an influx of intestinal Eomes+ T cells; by contrast, epithelial NKp44+CD69+ NK cells with less cytotoxic capacity persist in adults. In conclusion, NK cells are abundant in infant intestines, where they can provide effector functions while Eomes+ T cell responses mature. Innate lymphoid cells (ILC), including natural killer (NK) cells, are important innate immune regulators. Here the authors show that, in human infant intestines, CD103+Eomes+ NK cells are the predominant ILC population, but are replaced gradually by Eomes+ T cells, while NKp44+ NK cells persist in adult intestines.
Collapse
Affiliation(s)
- Adrian F Sagebiel
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251, Hamburg, Germany
| | - Fenja Steinert
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251, Hamburg, Germany
| | - Sebastian Lunemann
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251, Hamburg, Germany
| | - Christian Körner
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251, Hamburg, Germany
| | - Renée R C E Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Marcus Altfeld
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251, Hamburg, Germany
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Madeleine J Bunders
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251, Hamburg, Germany. .,Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands. .,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
45
|
Caudana P, Núñez NG, De La Rochere P, Pinto A, Denizeau J, Alonso R, Niborski LL, Lantz O, Sedlik C, Piaggio E. IL2/Anti-IL2 Complex Combined with CTLA-4, But Not PD-1, Blockade Rescues Antitumor NK Cell Function by Regulatory T-cell Modulation. Cancer Immunol Res 2019; 7:443-457. [PMID: 30651291 DOI: 10.1158/2326-6066.cir-18-0697] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/21/2018] [Accepted: 01/11/2019] [Indexed: 11/16/2022]
Abstract
High-dose IL2 immunotherapy can induce long-lasting cancer regression but is toxic and insufficiently efficacious. Improvements are obtained with IL2/anti-IL2 complexes (IL2Cx), which redirect IL2 action to CD8+ T and natural killer (NK) cells. Here, we evaluated the efficacy of combining IL2Cx with blockade of inhibitory immune pathways. In an autochthonous lung adenocarcinoma model, we show that the IL2Cx/anti-PD-1 combination increases CD8+ T-cell infiltration of the lung and controls tumor growth. In the B16-OVA model, which is resistant to checkpoint inhibition, combination of IL2Cx with PD-1 or CTLA-4 pathway blockade reverses that resistance. Both combinations work by reinvigorating exhausted intratumoral CD8+ T cells and by increasing the breadth of tumor-specific T-cell responses. However, only the IL2Cx/anti-CTLA-4 combination is able to rescue NK cell antitumor function by modulating intratumoral regulatory T cells. Overall, association of IL2Cx with PD-1 or CTLA-4 pathway blockade acts by different cellular mechanisms, paving the way for the rational design of combinatorial antitumor therapies.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antigen-Antibody Complex/therapeutic use
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen/immunology
- Cell Line, Tumor
- Disease Models, Animal
- Drug Resistance, Neoplasm/drug effects
- Female
- Immunotherapy
- Interleukin-2/immunology
- Killer Cells, Natural/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Programmed Cell Death 1 Receptor/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Pamela Caudana
- Institut Curie, PSL Research University, INSERM U932, TransImm Team, Paris, France
| | | | | | - Anaïs Pinto
- Institut Curie, PSL Research University, INSERM U932, TransImm Team, Paris, France
| | - Jordan Denizeau
- Institut Curie, PSL Research University, INSERM U932, TransImm Team, Paris, France
| | - Ruby Alonso
- Institut Curie, PSL Research University, INSERM U932, TransImm Team, Paris, France
| | | | - Olivier Lantz
- Institut Curie, PSL Research University, INSERM U932, TransImm Team, Paris, France
- Institut Curie, PSL Research University, Clinical Immunology Laboratory, Paris, France
- Centre d'Investigation Clinique Biothérapie CICBT 1428, Institut Curie, Paris, France
| | - Christine Sedlik
- Institut Curie, PSL Research University, INSERM U932, TransImm Team, Paris, France
| | - Eliane Piaggio
- Institut Curie, PSL Research University, INSERM U932, TransImm Team, Paris, France.
- Centre d'Investigation Clinique Biothérapie CICBT 1428, Institut Curie, Paris, France
| |
Collapse
|
46
|
Ma Q, Long W, Xing C, Chu J, Luo M, Wang HY, Liu Q, Wang RF. Cancer Stem Cells and Immunosuppressive Microenvironment in Glioma. Front Immunol 2018; 9:2924. [PMID: 30619286 PMCID: PMC6308128 DOI: 10.3389/fimmu.2018.02924] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022] Open
Abstract
Glioma is one of the most common malignant tumors of the central nervous system and is characterized by extensive infiltrative growth, neovascularization, and resistance to various combined therapies. In addition to heterogenous populations of tumor cells, the glioma stem cells (GSCs) and other nontumor cells present in the glioma microenvironment serve as critical regulators of tumor progression and recurrence. In this review, we discuss the role of several resident or peripheral factors with distinct tumor-promoting features and their dynamic interactions in the development of glioma. Localized antitumor factors could be silenced or even converted to suppressive phenotypes, due to stemness-related cell reprogramming and immunosuppressive mediators in glioma-derived microenvironment. Furthermore, we summarize the latest knowledge on GSCs and key microenvironment components, and discuss the emerging immunotherapeutic strategies to cure this disease.
Collapse
Affiliation(s)
- Qianquan Ma
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Wenyong Long
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Changsheng Xing
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Junjun Chu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Mei Luo
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Helen Y Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Qing Liu
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Rong-Fu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, United States.,Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, United States
| |
Collapse
|
47
|
Oyer JL, Gitto SB, Altomare DA, Copik AJ. PD-L1 blockade enhances anti-tumor efficacy of NK cells. Oncoimmunology 2018; 7:e1509819. [PMID: 30377572 PMCID: PMC6205063 DOI: 10.1080/2162402x.2018.1509819] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 08/01/2018] [Accepted: 08/04/2018] [Indexed: 12/31/2022] Open
Abstract
Anti-PD-1/anti-PD-L1 therapies have shown success in cancer treatment but responses are limited to ~ 15% of patients with lymphocyte infiltrated, PD-L1 positive tumors. Hence, strategies that increase PD-L1 expression and tumor infiltration should make more patients eligible for PD-1/PD-L1 blockade therapy, thus improving overall outcomes. PD-L1 expression on tumors is induced by IFNγ, a cytokine secreted by NK cells. Therefore, we tested if PM21-particle expanded NK cells (PM21-NK cells) induced expression of PD-L1 on tumors and if anti-PD-L1 treatment enhanced NK cell anti-tumor efficacy in an ovarian cancer model. Studies here showed that PM21-NK cells secrete high amounts of IFNγ and that adoptively transferred PM21-NK cells induce PD-L1 expression on SKOV-3 cells in vivo. The induction of PD-L1 expression on SKOV-3 cells coincided with the presence of regulatory T cells (Tregs) in the abdominal cavity and within tumors. In in vitro experiments, anti-PD-L1 treatment had no direct effect on cytotoxicity or cytokine secretion by predominantly PD-1 negative PM21-NK cells in response to PD-L1+ targets. However, significant improvement of NK cell anti-tumor efficacy was observed in vivo when combined with anti-PD-L1. PD-L1 blockade also resulted in increased in vivo NK cell persistence and retention of their cytotoxic phenotype. These results support the use of anti-PD-L1 in combination with NK cell therapy regardless of initial tumor PD-L1 status and indicate that NK cell therapy would likely augment the applicability of anti-PD-L1 treatment.
Collapse
Affiliation(s)
- Jeremiah L Oyer
- Burnett School of Biomedical Sciences, University of Central Florida, College of Medicine, Orlando, FL, USA
| | - Sarah B Gitto
- Burnett School of Biomedical Sciences, University of Central Florida, College of Medicine, Orlando, FL, USA
| | - Deborah A Altomare
- Burnett School of Biomedical Sciences, University of Central Florida, College of Medicine, Orlando, FL, USA
| | - Alicja J Copik
- Burnett School of Biomedical Sciences, University of Central Florida, College of Medicine, Orlando, FL, USA
| |
Collapse
|
48
|
Fleischer LM, Somaiya RD, Miller GM. Review and Meta-Analyses of TAAR1 Expression in the Immune System and Cancers. Front Pharmacol 2018; 9:683. [PMID: 29997511 PMCID: PMC6029583 DOI: 10.3389/fphar.2018.00683] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 06/06/2018] [Indexed: 12/29/2022] Open
Abstract
Since its discovery in 2001, the major focus of TAAR1 research has been on its role in monoaminergic regulation, drug-induced reward and psychiatric conditions. More recently, TAAR1 expression and functionality in immune system regulation and immune cell activation has become a topic of emerging interest. Here, we review the immunologically-relevant TAAR1 literature and incorporate open-source expression and cancer survival data meta-analyses. We provide strong evidence for TAAR1 expression in the immune system and cancers revealed through NCBI GEO datamining and discuss its regulation in a spectrum of immune cell types as well as in numerous cancers. We discuss connections and logical directions for further study of TAAR1 in immunological function, and its potential role as a mediator or modulator of immune dysregulation, immunological effects of psychostimulant drugs of abuse, and cancer progression.
Collapse
Affiliation(s)
- Lisa M Fleischer
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Rachana D Somaiya
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Gregory M Miller
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States.,Department of Chemical Engineering, Northeastern University, Boston, MA, United States.,Center for Drug Discovery, Northeastern University, Boston, MA, United States
| |
Collapse
|
49
|
Babic M, Romagnani C. The Role of Natural Killer Group 2, Member D in Chronic Inflammation and Autoimmunity. Front Immunol 2018; 9:1219. [PMID: 29910814 PMCID: PMC5992374 DOI: 10.3389/fimmu.2018.01219] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/15/2018] [Indexed: 01/06/2023] Open
Abstract
Current medicine and medical science puts great effort into elucidating the basis of chronicity and finding appropriate treatments for inflammatory diseases; however, the mechanisms driving aberrant immune responses are mostly unknown and deserve further study. Of particular interest is the identification of checkpoints that regulate the function and differentiation of pro-inflammatory cells during pathogenesis, along with means of their modulation for therapeutic purposes. Natural killer group 2, member D (NKG2D) is a potent activator of the immune system, known as a sensor for “induced-self” ligands, i.e., cellular danger signals that, in the context of chronic inflammation and autoimmunity, can be presented by cells being exposed to an inflammatory cytokine milieu, endoplasmic reticulum stress, or cell death. Engagement by such ligands can be translated by NKG2D into activation or co-stimulation of NK cells and different subsets of T cells, respectively, thus contributing to the regulation of the inflammatory response. In this review, we discuss the current knowledge on the contribution of the NKG2D–NKG2DL signaling axis during intestinal inflammation, type 1 diabetes, multiple sclerosis, and rheumatoid arthritis, where the role of NKG2D has been associated either by aberrant expression of the receptor and its ligands and/or by functional data in corresponding mouse models.
Collapse
Affiliation(s)
- Marina Babic
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany.,Medical Department I, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Chiara Romagnani
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany.,Medical Department I, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
50
|
Jeffery HC, Braitch MK, Bagnall C, Hodson J, Jeffery LE, Wawman RE, Wong LL, Birtwistle J, Bartlett H, Lohse AW, Hirschfield GM, Dyson J, Jones D, Hubscher SG, Klenerman P, Adams DH, Oo YH. Changes in natural killer cells and exhausted memory regulatory T Cells with corticosteroid therapy in acute autoimmune hepatitis. Hepatol Commun 2018; 2:421-436. [PMID: 29619420 PMCID: PMC5880196 DOI: 10.1002/hep4.1163] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 12/24/2022] Open
Abstract
Autoimmune hepatitis (AIH) is an immune-mediated liver disease currently treated by immunosuppressive medications with significant side effects. Thus, novel mechanistic treatments are greatly needed. We performed prospective deep immunophenotyping of blood immune cells in patients with acute AIH before and after corticosteroid therapy. Blood samples from 26 patients with acute AIH (United Kingdom-AIH Consortium) were phenotyped by flow cytometry at baseline and 4 months after starting corticosteroids. Pretreatment liver tissues were stained for forkhead box P3-positive (FOXP3POS) regulatory T cells (Tregs), clusters of differentiation (CD)56POS natural killer (NK) cells, and chemokine (C-X-C motif) ligand 10. Chemokine secretion by cultured primary hepatocyte and biliary epithelial cells was measured by enzyme-linked immunosorbent assay. Functional coculture assays with stimulated NK cells and Tregs were performed. CD161 ligand, lectin-like transcript-1 expression by intrahepatic immune cells was demonstrated with flow cytometry. Frequencies of NKbright cells declined with therapy (P < 0.001) and correlated with levels of alanine aminotransferase (P = 0.023). The Treg:NKbright ratio was lower pretreatment, and Tregs had an activated memory phenotype with high levels of CD39, cytotoxic T lymphocyte antigen 4, and FOXP3 but also high programmed death ligand 1, indicating exhaustion. Coculture experiments suggested the Tregs could not efficiently suppress interferon-γ secretion by NK cells. Both Tregs and NK cells had high expression of liver infiltration and T helper 17 plasticity-associated marker CD161 (P = 0.04). Pretreatment and CD161pos NK cells expressed high levels of perforin and granzyme B, consistent with an activated effector phenotype (P < 0.05). Lectin-like transcript 1, a ligand for CD161, is expressed on intrahepatic B cells, monocytes, and neutrophils. Conclusion: Activated effector NK cells, which correlate with biochemical measurements of hepatitis, and exhausted memory Tregs are increased in the blood of patients with treatment-naive AIH and decline with corticosteroid therapy. Inadequate regulation of NK cells by exhausted FOXP3pos Tregs may play a role in AIH pathogenesis and contribute to liver injury. (Hepatology Communications 2018;2:421-436).
Collapse
Affiliation(s)
- Hannah C. Jeffery
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
| | - Manjit K. Braitch
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
| | - Chris Bagnall
- Human Biomaterials Resource CentreUniversity of BirminghamUnited Kingdom
| | - James Hodson
- Institute of Translational MedicineUniversity Hospitals Birmingham National Health Services Foundation Trust, University of BirminghamBirminghamUnited Kingdom
| | - Louisa E. Jeffery
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
| | - Rebecca E. Wawman
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- School of Life Sciences, Faculty of Health and Life SciencesCoventry UniversityCoventryUnited Kingdom
| | - Lin Lee Wong
- Newcastle Biomedical Research Centre and Newcastle UniversityNewcastleUnited Kingdom
| | - Jane Birtwistle
- Clinical Immunology DepartmentUniversity of BirminghamBirminghamUnited Kingdom
| | - Helen Bartlett
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
| | | | - Gideon M. Hirschfield
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- Liver Transplantation and Hepatobiliary Unit, Queen Elizabeth HospitalUniversity Hospitals Birmingham National Health Service Foundation TrustBirminghamUnited Kingdom
| | - Jessica Dyson
- Newcastle Biomedical Research Centre and Newcastle UniversityNewcastleUnited Kingdom
| | - David Jones
- Newcastle Biomedical Research Centre and Newcastle UniversityNewcastleUnited Kingdom
| | - Stefan G. Hubscher
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- Department of Histopathology, Queen Elizabeth HospitalUniversity Hospitals Birmingham National Health Service Foundation TrustBirminghamUnited Kingdom
| | - Paul Klenerman
- Peter Medawar Building of Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom
| | - David H. Adams
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- Liver Transplantation and Hepatobiliary Unit, Queen Elizabeth HospitalUniversity Hospitals Birmingham National Health Service Foundation TrustBirminghamUnited Kingdom
| | - Ye H. Oo
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- Liver Transplantation and Hepatobiliary Unit, Queen Elizabeth HospitalUniversity Hospitals Birmingham National Health Service Foundation TrustBirminghamUnited Kingdom
| |
Collapse
|