1
|
Wu TN, Hung JT, Hung TH, Wang YH, Wu JC, Yu AL. Effective suppression of tumor growth and hepatic metastasis of neuroblastoma by NKT-stimulatory phenyl glycolipid. Biomed Pharmacother 2024; 177:117040. [PMID: 38959605 DOI: 10.1016/j.biopha.2024.117040] [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: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Invariant natural killer T cell (iNKT) cells produce large amounts of cytokines in response to α-Galactosylceramide (α-GalCer) stimulation. An analog containing two phenyl rings on the acyl chain, C34, was previously found to be more Th1-biased than α-GalCer and triggered greater anticancer activities against breast cancer, melanoma and lung cancer in mice. Since liver is enriched in iNKT cells, we investigated anticancer efficacy of C34 on neuroblastoma with hepatic metastasis. C34 induced Th1-biased cytokine secretions in the liver, significantly suppressed neuroblastoma growth/metastasis and prolonged mouse survival. The anti-tumor efficacy might be attributed to greater expansions of hepatic NKT, NK, CD4+ T, and CD8+ T cells as well as reduction of the number of SSCloGr1intCD11b+ subset of myeloid-derived suppressor cells (MDSCs) in the liver of tumor-bearing mice, as compared to DMSO control group. C34 also upregulated expression of CD1d and CD11c, especially in the SSCloGr1intCD11b+ subset of MDSCs, which might be killed by C34-activated NKT cells, attributing to their reduced number. In addition, C34 also induced expansion of CD4+ T, CD8+ T, and NK cells, which might eliminate neuroblastoma cells. These immune-modulating effects of C34 might act in concert in the local milieu of liver to suppress the tumor growth. Further analysis of database of neuroblastoma revealed that patients with high CD11c expression in the monocytic MDSCs in the tumor had longer survival, suggesting the potential clinical application of C34 for treatment of neuroblastoma.
Collapse
Affiliation(s)
- Tai-Na Wu
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; Institute of Biotechnology, National Taiwan University, Taipei 115, Taiwan.
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan.
| | - Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Hui Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Jen-Chine Wu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan; Department of Pediatrics, University of California in San Diego, La Jolla, CA, USA
| |
Collapse
|
2
|
Metelitsa LS. A milestone method to make natural killer T cells. Nat Biotechnol 2024:10.1038/s41587-024-02243-x. [PMID: 38744945 DOI: 10.1038/s41587-024-02243-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Affiliation(s)
- Leonid S Metelitsa
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer and Hematology Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Fujii SI, Shimizu K. NKT-Licensed In Vivo Dendritic Cell-Based Immunotherapy as Cellular Immunodrugs for Cancer Treatment. Crit Rev Oncog 2024; 29:45-61. [PMID: 38421713 DOI: 10.1615/critrevoncog.2023048735] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
With the advent of new therapies, immunotherapy has gained attention as a critical modality. After the discovery of the natural killer T (NKT) cells ligand, ex vivo cultured dendritic cells (DCs) loaded with NKT ligand (especially α-galactosylceramide (α-GalCer) (DC/Gal) or ex vivo expanded NKT transfer studies were clinically examined in several institutes. To prevent tumoral immune escape, the link between innate and adaptive immunity, in situ selective targeting of DCs has been attempted; however, protocol optimization was required. As a type of DC targeting therapy that combines the benefits of invariant natural killer T (iNKT) cells, we established an all-in-one, off-the-shelf drug, named the artificial adjuvant vector cell (aAVC), which consists of the tumor antigen and the CD1d-iNKT ligand complex. Here, to our knowledge, we first demonstrate the DC/GalCer therapy and NKT transfer therapy. Next, we introduce and discuss the use of aAVC therapy not only for efficient innate and adaptive immunity induction using fully matured DC in situ but also the characterization necessary for locally reprogramming the tumor microenvironment and systemically inducing long-term memory in T cells. We also discuss how the immune network mechanism is controlled by DCs. Next, we performed the first human clinical trial using WT1 antigen-expressing aAVC against relapse and refractory acute myelogenous leukemia. Thus, we highlight the challenges of using aAVCs as prodrugs for actively energizing DCs in vivo, underpinning immunological networks, and developing strategies for providing maximal benefits for patients.
Collapse
Affiliation(s)
- Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), and RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama, Kanagawa, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), and RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama, Kanagawa, Japan
| |
Collapse
|
4
|
Burn OK, Dasyam N, Hermans IF. Recruiting Natural Killer T Cells to Improve Vaccination: Lessons from Preclinical and Clinical Studies. Crit Rev Oncog 2024; 29:31-43. [PMID: 38421712 DOI: 10.1615/critrevoncog.2023049407] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The capacity of type I natural killer T (NKT) cells to provide stimulatory signals to antigen-presenting cells has prompted preclinical research into the use of agonists as immune adjuvants, with much of this work focussed on stimulating T cell responses to cancer. In attempting to evaluate this approach in the clinic, our recent dendritic-cell based study failed to show an advantage to adding an agonist to the vaccine. Here we present potential limitations of the study, and suggest why other simpler strategies may be more effective. These include strategies to target antigen-presenting cells in the host, either through promoting efficient transfer from injected cell lines, facilitating uptake of antigen and agonist as injected conjugates, or encapsulating the components into injected nanovectors. While the vaccine landscape has changed with the rapid uptake of mRNA vaccines, we suggest that there is still a role for recruiting NKT cells in altering T cell differentiation programmes, notably the induction of resident memory T cells.
Collapse
Affiliation(s)
- Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
5
|
Gonzatti MB, Freire BM, Antunes MM, de Menezes GB, Talbot J, Peron JPS, Basso AS, Keller AC. Targeting adrenergic receptors to mitigate invariant natural killer T cells-induced acute liver injury. iScience 2023; 26:107947. [PMID: 37841583 PMCID: PMC10568435 DOI: 10.1016/j.isci.2023.107947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/07/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Invariant Natural Killer T (iNKT) cell activation by α-galactosylceramide (αGC) potentiates cytotoxic immune responses against tumors. However, αGC-induced liver injury is a limiting factor for iNKT-based immunotherapy. Although adrenergic receptor stimulation is an important immunosuppressive signal that curbs tissue damage induced by inflammation, its effect on the antitumor activity of invariant Natural Killer T (iNKT) cells remains unclear. We use mouse models and pharmacological tools to show that the stimulation of the sympathetic nervous system (SNS) inhibits αGC-induced liver injury without impairing iNKT cells' antitumoral functions. Mechanistically, SNS stimulation prevents the collateral effect of TNF-α production by iNKT cells and neutrophil accumulation in hepatic parenchyma. Our results suggest that the modulation of the adrenergic signaling can be a complementary approach to αGC-based immunotherapy to mitigate iNKT-induced liver injury without compromising its antitumoral activity.
Collapse
Affiliation(s)
- Michelangelo Bauwelz Gonzatti
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), Rua Botucatu, 862, 4th floor, São Paulo 04023-062, Brazil
| | - Beatriz Marton Freire
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), Rua Botucatu, 862, 4th floor, São Paulo 04023-062, Brazil
| | - Maísa Mota Antunes
- Department of Morphology, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Minas Gerais 31270-910, Brazil
| | - Gustavo Batista de Menezes
- Department of Morphology, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Minas Gerais 31270-910, Brazil
| | - Jhimmy Talbot
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA 98109-1024, USA
| | - Jean Pierre Schatzmann Peron
- Department of Immunology-ICB IV, University of São Paulo, Av. Prof. Lineu Prestes, 1730, São Paulo 05508-900, Brazil
| | - Alexandre Salgado Basso
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), Rua Botucatu, 862, 4th floor, São Paulo 04023-062, Brazil
| | - Alexandre Castro Keller
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), Rua Botucatu, 862, 4th floor, São Paulo 04023-062, Brazil
| |
Collapse
|
6
|
Zheng Z, Wieder T, Mauerer B, Schäfer L, Kesselring R, Braumüller H. T Cells in Colorectal Cancer: Unravelling the Function of Different T Cell Subsets in the Tumor Microenvironment. Int J Mol Sci 2023; 24:11673. [PMID: 37511431 PMCID: PMC10380781 DOI: 10.3390/ijms241411673] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Therapeutic options for metastatic colorectal cancer (mCRC) are very limited, and the prognosis using combination therapy with a chemotherapeutic drug and a targeted agent, e.g., epidermal growth factor receptor or tyrosine kinase, remains poor. Therefore, mCRC is associated with a poor median overall survival (mOS) of only 25-30 months. Current immunotherapies with checkpoint inhibitor blockade (ICB) have led to a substantial change in the treatment of several cancers, such as melanoma and non-small cell lung cancer. In CRC, ICB has only limited effects, except in patients with microsatellite instability-high (MSI-H) or mismatch repair-deficient (dMMR) tumors, which comprise about 15% of sporadic CRC patients and about 4% of patients with metastatic CRC. The vast majority of sporadic CRCs are microsatellite-stable (MSS) tumors with low levels of infiltrating immune cells, in which immunotherapy has no clinical benefit so far. Immunotherapy with checkpoint inhibitors requires the presence of infiltrating T cells into the tumor microenvironment (TME). This makes T cells the most important effector cells in the TME, as evidenced by the establishment of the immunoscore-a method to estimate the prognosis of CRC patients. The microenvironment of a tumor contains several types of T cells that are anti-tumorigenic, such as CD8+ T cells or pro-tumorigenic, such as regulatory T cells (Tregs) or T helper 17 (Th17) cells. However, even CD8+ T cells show marked heterogeneity, e.g., they can become exhausted, enter a state of hyporesponsiveness or become dysfunctional and express high levels of checkpoint molecules, the targets for ICB. To kill cancer cells, CD8+ T cells need the recognition of the MHC class I, which is often downregulated on colorectal cancer cells. In this case, a population of unconventional T cells with a γδ T cell receptor can overcome the limitations of the conventional CD8+ T cells with an αβT cell receptor. γδ T cells recognize antigens in an MHC-independent manner, thus acting as a bridge between innate and adaptive immunity. Here, we discuss the effects of different T cell subsets in colorectal cancer with a special emphasis on γδ T cells and the possibility of using them in CAR-T cell therapy. We explain T cell exclusion in microsatellite-stable colorectal cancer and the possibilities to overcome this exclusion to enable immunotherapy even in these "cold" tumors.
Collapse
Affiliation(s)
- Ziwen Zheng
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Thomas Wieder
- Department of Vegetative and Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Bernhard Mauerer
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Luisa Schäfer
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Rebecca Kesselring
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Heidi Braumüller
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
7
|
You M, Liu J, Li J, Ji C, Ni H, Guo W, Zhang J, Jia W, Wang Z, Zhang Y, Yao Y, Yu G, Ji H, Wang X, Han D, Du X, Xu MM, Yu S. Mettl3-m 6A-Creb1 forms an intrinsic regulatory axis in maintaining iNKT cell pool and functional differentiation. Cell Rep 2023; 42:112584. [PMID: 37267102 DOI: 10.1016/j.celrep.2023.112584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/07/2023] [Accepted: 05/15/2023] [Indexed: 06/04/2023] Open
Abstract
N6-methyladenosine (m6A) methyltransferase Mettl3 is involved in conventional T cell immunity; however, its role in innate immune cells remains largely unknown. Here, we show that Mettl3 intrinsically regulates invariant natural killer T (iNKT) cell development and function in an m6A-dependent manner. Conditional ablation of Mettl3 in CD4+CD8+ double-positive (DP) thymocytes impairs iNKT cell proliferation, differentiation, and cytokine secretion, which synergistically causes defects in B16F10 melanoma resistance. Transcriptomic and epi-transcriptomic analyses reveal that Mettl3 deficiency disturbs the expression of iNKT cell-related genes with altered m6A modification. Strikingly, Mettl3 modulates the stability of the Creb1 transcript, which in turn controls the protein and phosphorylation levels of Creb1. Furthermore, conditional targeting of Creb1 in DP thymocytes results in similar phenotypes of iNKT cells lacking Mettl3. Importantly, ectopic expression of Creb1 largely rectifies such developmental defects in Mettl3-deficient iNKT cells. These findings reveal that the Mettl3-m6A-Creb1 axis plays critical roles in regulating iNKT cells at the post-transcriptional layer.
Collapse
Affiliation(s)
- Menghao You
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jingjing Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jie Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China; Department of Basic Medical Sciences, School of Medicine, Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, THU-PKU Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ce Ji
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Haochen Ni
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
| | - Wenhui Guo
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiarui Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Weiwei Jia
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Zhao Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yajiao Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yingpeng Yao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guotao Yu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Huanyu Ji
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xiaohu Wang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dali Han
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
| | - Xuguang Du
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Meng Michelle Xu
- Department of Basic Medical Sciences, School of Medicine, Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, THU-PKU Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Shuyang Yu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
8
|
Yi Q, Wang J, Liu T, Yao Y, Loveless I, Subedi K, Toor J, Adrianto I, Xiao H, Chen B, Crawford HC, Fang D, Zhou L, Mi QS. scRNA-Seq and imaging mass cytometry analyses unveil iNKT cells-mediated anti-tumor immunity in pancreatic cancer liver metastasis. Cancer Lett 2023; 561:216149. [PMID: 36990268 PMCID: PMC11737350 DOI: 10.1016/j.canlet.2023.216149] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/18/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
Invariant natural killer T (iNKT) cells are innate-like T cells that are abundant in liver sinusoids and play a critical role in tumor immunity. However, the role of iNKT cells in pancreatic cancer liver metastasis (PCLM) has not been fully explored. In this study, we employed a hemi-spleen pancreatic tumor cell injection mouse model of PCLM, a model that closely mimics clinical conditions in humans, to explore the role of iNKT cells in PCLM. Activation of iNKT cells with α-galactosylceramide (αGC) markedly increased immune cell infiltration and suppressed PCLM progression. Via single cell RNA sequencing (scRNA-seq) we profiled over 30,000 immune cells from normal liver and PCLM with or without αGC treatment and were able to characterize the global changes of the immune cells in the tumor microenvironment upon αGC treatment, identifying a total of 12 subpopulations. Upon treatment with αGC, scRNA-Seq and flow cytometry analyses revealed increased cytotoxic activity of iNKT/NK cells and skewing CD4 T cells towards a cytotoxic Th1 profile and CD8 T cells towards a cytotoxic profile, characterized by higher proliferation and reduced exhaustion marker PD1 expression. Moreover, αGC treatment excluded tumor associated macrophages. Lastly, imaging mass cytometry analysis uncovered the reduced epithelial to mesenchymal transition related markers and increased active CD4 and CD8 T cells in PCLM with αGC treatment. Overall, our findings uncover the protective function of activated iNKT cells in pancreatic cancer liver metastasis through increased NK and T cell immunity and decreased tumor associated macrophages.
Collapse
Affiliation(s)
- Qijun Yi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Jie Wang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Tingting Liu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Yi Yao
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Ian Loveless
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA; Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, 48202, USA
| | - Kalpana Subedi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Jugmohit Toor
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Indra Adrianto
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA; Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, 48202, USA; Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Hua Xiao
- Department of Physiology, College of Natural Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Bin Chen
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, USA
| | - Howard C Crawford
- Henry Ford Pancreatic Cancer Center, Department of Surgery, Henry Ford Health, Detroit, MI, 48202, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA; Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, 48202, USA; Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA; Department of Internal Medicine, Henry Ford Health, Detroit, MI, 48202, USA.
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA; Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, 48202, USA; Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA; Department of Internal Medicine, Henry Ford Health, Detroit, MI, 48202, USA.
| |
Collapse
|
9
|
Ruf B, Greten TF, Korangy F. Innate lymphoid cells and innate-like T cells in cancer - at the crossroads of innate and adaptive immunity. Nat Rev Cancer 2023; 23:351-371. [PMID: 37081117 DOI: 10.1038/s41568-023-00562-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 04/22/2023]
Abstract
Immunotherapies targeting conventional T cells have revolutionized systemic treatment for many cancers, yet only a subset of patients benefit from these approaches. A better understanding of the complex immune microenvironment of tumours is needed to design the next generation of immunotherapeutics. Innate lymphoid cells (ILCs) and innate-like T cells (ILTCs) are abundant, tissue-resident lymphocytes that have recently been shown to have critical roles in many types of cancers. ILCs and ILTCs rapidly respond to changes in their surrounding environment and act as the first responders to bridge innate and adaptive immunity. This places ILCs and ILTCs as pivotal orchestrators of the final antitumour immune response. In this Review, we outline hallmarks of ILCs and ILTCs and discuss their emerging role in antitumour immunity, as well as the pathophysiological adaptations leading to their pro-tumorigenic function. We explore the pleiotropic, in parts redundant and sometimes opposing, mechanisms that underlie the delicate interplay between the different subsets of ILCs and ILTCs. Finally, we highlight their role in amplifying and complementing conventional T cell functions and summarize immunotherapeutic strategies for targeting ILCs and ILTCs in cancer.
Collapse
Affiliation(s)
- Benjamin Ruf
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI CCR Liver Cancer Program, National Institutes of Health, Bethesda, MD, USA
| | - Firouzeh Korangy
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
10
|
Look A, Burns D, Tews I, Roghanian A, Mansour S. Towards a better understanding of human iNKT cell subpopulations for improved clinical outcomes. Front Immunol 2023; 14:1176724. [PMID: 37153585 PMCID: PMC10154573 DOI: 10.3389/fimmu.2023.1176724] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a unique T lymphocyte population expressing semi-invariant T cell receptors (TCRs) that recognise lipid antigens presented by CD1d. iNKT cells exhibit potent anti-tumour activity through direct killing mechanisms and indirectly through triggering the activation of other anti-tumour immune cells. Because of their ability to induce potent anti-tumour responses, particularly when activated by the strong iNKT agonist αGalCer, they have been the subject of intense research to harness iNKT cell-targeted immunotherapies for cancer treatment. However, despite potent anti-tumour efficacy in pre-clinical models, the translation of iNKT cell immunotherapy into human cancer patients has been less successful. This review provides an overview of iNKT cell biology and why they are of interest within the context of cancer immunology. We focus on the iNKT anti-tumour response, the seminal studies that first reported iNKT cytotoxicity, their anti-tumour mechanisms, and the various described subsets within the iNKT cell repertoire. Finally, we discuss several barriers to the successful utilisation of iNKT cells in human cancer immunotherapy, what is required for a better understanding of human iNKT cells, and the future perspectives facilitating their exploitation for improved clinical outcomes.
Collapse
Affiliation(s)
- Alex Look
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Daniel Burns
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ivo Tews
- Biological Sciences, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Ali Roghanian
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Salah Mansour
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
11
|
Courtney AN, Tian G, Metelitsa LS. Natural killer T cells and other innate-like T lymphocytes as emerging platforms for allogeneic cancer cell therapy. Blood 2023; 141:869-876. [PMID: 36347021 PMCID: PMC10023720 DOI: 10.1182/blood.2022016201] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/19/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
T cells expressing chimeric antigen receptors (CARs) have achieved major clinical success in patients with hematologic malignancies. However, these treatments remain largely ineffective for solid cancers and require significant time and resources to be manufactured in an autologous setting. Developing alternative immune effector cells as cancer immunotherapy agents that can be employed in allogeneic settings is crucial for the advancement of cell therapy. Unlike T cells, Vα24-invariant natural killer T cells (NKTs) are not alloreactive and can therefore be generated from allogeneic donors for rapid infusion into numerous patients without the risk of graft-versus-host disease. Additionally, NKT cells demonstrate inherent advantages over T-cell products, including the ability to traffic to tumor tissues, target tumor-associated macrophages, transactivate NK cells, and cross-prime tumor-specific CD8 T cells. Both unmodified NKTs, which specifically recognize CD1d-bound glycolipid antigens expressed by certain types of tumors, and CAR-redirected NKTs are being developed as the next generation of allogeneic cell therapy products. In this review, we describe studies on the biology of NKTs and other types of innate-like T cells and summarize the clinical experiences of unmodified and CAR-redirected NKTs, including recent interim reports on allogeneic NKTs.
Collapse
Affiliation(s)
- Amy N. Courtney
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX
| | - Gengwen Tian
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX
| | - Leonid S. Metelitsa
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| |
Collapse
|
12
|
Iyoda T, Shimizu K, Kawamura M, Shinga J, Watanabe T, Fukunaga K, Mushiroda T, Saka H, Kitagawa C, Shimamatsu SI, Takenoyama M, Suehiro Y, Imai T, Shintani A, Ito S, Fujii SI. Augmenting Granzyme B-Expressing NK Cells by Invariant NKT Ligand-Loaded APCs in Patients with Postoperative Early Stage Non-Small Cell Lung Cancer: Results of a Randomized Phase II Study. Immunohorizons 2023; 7:1-16. [PMID: 36637516 PMCID: PMC10563390 DOI: 10.4049/immunohorizons.2200091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 01/14/2023] Open
Abstract
NK cells are major effector cells involved in the elimination of early tumors and prevent metastasis. They often have an impaired function in patients with cancer. Preclinical studies have demonstrated NK cell activation as the adjunctive effect of invariant NKT (iNKT) cells. Activation of iNKT cells after administration of the glycolipid ligand α-galactosylceramide, loaded with CD1d-expressing human PBMC-derived APCs (APC/Gal), is an attractive cancer therapy to optimize the use of NK cells. However, the subsets of NK cells that are activated following iNKT cell activation as well as the period of NK cell activation remain unclear. In this study, we report that the granzyme B-expressing NK cell response in postoperative lung cancer patients was enhanced 49 d after administration of APC/Gal in a phase II study. We found maximum IFN-γ production on day 49 in 13 out of 27 APC/Gal-treated patients. On day 49, 14 out of 27 patients (51.9%) had higher IFN-γ production by iNKT cells (>6-fold higher than the baseline level). This increment significantly correlated with granzyme B-expressing NK cells. Although IFN-γ production was lower in patients in the nontreated group, we detected maximum IFN-γ production 12 mo after the resection of lung cancer (9 out of 29 patients [31%]). These findings suggest that elimination of cancer cells leads to increased NK cell function, which can be further enhanced by APC/Gal therapy.
Collapse
Affiliation(s)
- Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- RIKEN Drug Discovery and Medical Technology Platforms, Yokohama, Japan
| | - Masami Kawamura
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Jun Shinga
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takashi Watanabe
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Koya Fukunaga
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Taisei Mushiroda
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hideo Saka
- Department of Respiratory Medicine, Nagoya Medical Center, National Hospital Organization, Nagoya, Japan
| | - Chiyoe Kitagawa
- Department of Medical Oncology, Nagoya Medical Center, National Hospital Organization, Nagoya, Japan
| | - Shin-ichiro Shimamatsu
- Department of Thoracic Oncology, Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - Mitsuhiro Takenoyama
- Department of Thoracic Oncology, Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - Youko Suehiro
- Department of Hematology, Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - Takumi Imai
- Department of Medical Statistics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; and
| | - Ayumi Shintani
- Department of Medical Statistics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; and
- Department of Clinical Research, Clinical Research Center, National Hospital Organization, Tokyo, Japan
| | - Suminobu Ito
- Department of Clinical Research, Clinical Research Center, National Hospital Organization, Tokyo, Japan
| | - Shin-ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- RIKEN Drug Discovery and Medical Technology Platforms, Yokohama, Japan
| |
Collapse
|
13
|
Lin Tan Z, Miyanaga K, Kitamoto Y, Yamamoto N. Levilactobacillus brevis surface layer protein B promotes liposome targeting to antigen-presenting cells in Peyer’s patches. Int J Pharm 2022; 622:121896. [DOI: 10.1016/j.ijpharm.2022.121896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/20/2022] [Accepted: 06/03/2022] [Indexed: 11/30/2022]
|
14
|
Chu J, Gao F, Yan M, Zhao S, Yan Z, Shi B, Liu Y. Natural killer cells: a promising immunotherapy for cancer. J Transl Med 2022; 20:240. [PMID: 35606854 PMCID: PMC9125849 DOI: 10.1186/s12967-022-03437-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022] Open
Abstract
As a promising alternative platform for cellular immunotherapy, natural killer cells (NK) have recently gained attention as an important type of innate immune regulatory cell. NK cells can rapidly kill multiple adjacent cancer cells through non-MHC-restrictive effects. Although tumors may develop multiple resistance mechanisms to endogenous NK cell attack, in vitro activation, expansion, and genetic modification of NK cells can greatly enhance their anti-tumor activity and give them the ability to overcome drug resistance. Some of these approaches have been translated into clinical applications, and clinical trials of NK cell infusion in patients with hematological malignancies and solid tumors have thus far yielded many encouraging clinical results. CAR-T cells have exhibited great success in treating hematological malignancies, but their drawbacks include high manufacturing costs and potentially fatal toxicity, such as cytokine release syndrome. To overcome these issues, CAR-NK cells were generated through genetic engineering and demonstrated significant clinical responses and lower adverse effects compared with CAR-T cell therapy. In this review, we summarize recent advances in NK cell immunotherapy, focusing on NK cell biology and function, the types of NK cell therapy, and clinical trials and future perspectives on NK cell therapy.
Collapse
Affiliation(s)
- Junfeng Chu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Fengcai Gao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Meimei Yan
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Shuang Zhao
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Zheng Yan
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Bian Shi
- Department of Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China.
| | - Yanyan Liu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
15
|
Delfanti G, Dellabona P, Casorati G, Fedeli M. Adoptive Immunotherapy With Engineered iNKT Cells to Target Cancer Cells and the Suppressive Microenvironment. Front Med (Lausanne) 2022; 9:897750. [PMID: 35615083 PMCID: PMC9125179 DOI: 10.3389/fmed.2022.897750] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Invariant Natural Killer T (iNKT) cells are T lymphocytes expressing a conserved semi-invariant TCR specific for lipid antigens (Ags) restricted for the monomorphic MHC class I-related molecule CD1d. iNKT cells infiltrate mouse and human tumors and play an important role in the immune surveillance against solid and hematological malignancies. Because of unique functional features, they are attractive platforms for adoptive cells immunotherapy of cancer compared to conventional T cells. iNKT cells can directly kill CD1d-expressing cancer cells, but also restrict immunosuppressive myelomonocytic populations in the tumor microenvironment (TME) via CD1d-cognate recognition, promoting anti-tumor responses irrespective of the CD1d expression by cancer cells. Moreover, iNKT cells can be adoptively transferred across MHC barriers without risk of alloreaction because CD1d molecules are identical in all individuals, in addition to their ability to suppress graft vs. host disease (GvHD) without impairing the anti-tumor responses. Within this functional framework, iNKT cells are successfully engineered to acquire a second antigen-specificity by expressing recombinant TCRs or Chimeric Antigen Receptor (CAR) specific for tumor-associated antigens, enabling the direct targeting of antigen-expressing cancer cells, while maintaining their CD1d-dependent functions. These new evidences support the exploitation of iNKT cells for donor unrestricted, and possibly off the shelf, adoptive cell therapies enabling the concurrent targeting of cancer cells and suppressive microenvironment.
Collapse
Affiliation(s)
- Gloria Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- *Correspondence: Gloria Delfanti
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Giulia Casorati
| | - Maya Fedeli
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Maya Fedeli
| |
Collapse
|
16
|
Du W, Frankel TL, Green M, Zou W. IFNγ signaling integrity in colorectal cancer immunity and immunotherapy. Cell Mol Immunol 2022; 19:23-32. [PMID: 34385592 PMCID: PMC8752802 DOI: 10.1038/s41423-021-00735-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
The majority of colorectal cancer patients are not responsive to immune checkpoint blockade (ICB). The interferon gamma (IFNγ) signaling pathway drives spontaneous and ICB-induced antitumor immunity. In this review, we summarize recent advances in the epigenetic, genetic, and functional integrity of the IFNγ signaling pathway in the colorectal cancer microenvironment and its immunological relevance in the therapeutic efficacy of and resistance to ICB. Moreover, we discuss how to target IFNγ signaling to inform novel clinical trials to treat patients with colorectal cancer.
Collapse
Affiliation(s)
- Wan Du
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Timothy L Frankel
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Michael Green
- Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Veterans Affairs Ann Arbor Healthcare System, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Graduate Programs in Immunology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Graduate Programs in Immunology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Tumor Biology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
17
|
Abstract
Gastrointestinal (GI) cancers represent a complex array of cancers that affect the digestive system. This includes liver, pancreatic, colon, rectal, anal, gastric, esophageal, intestinal and gallbladder cancer. Patients diagnosed with certain GI cancers typically have low survival rates, so new therapeutic approaches are needed. A potential approach is to harness the potent immunoregulatory properties of natural killer T (NKT) cells which are true T cells, not natural killer (NK) cells, that recognize lipid instead of peptide antigens presented by the non-classical major histocompatibility (MHC) molecule CD1d. The NKT cell subpopulation is known to play a vital role in tumor immunity by bridging innate and adaptive immune responses. In GI cancers, NKT cells can contribute to either antitumor or protumor immunity depending on the cytokine profile expressed and type of cancer. This review discusses the complexities of the role of NKT cells in liver, colon, pancreatic and gastric cancers with an emphasis on type I NKT cells.
Collapse
Affiliation(s)
- Julian Burks
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA,CONTACT Julian Burks National Cancer Institute, National Institute of Health, Building 41/Room D702, 41 Medlars Drive, Bethesda, Maryland20892, USA
| | - Purevdorj B. Olkhanud
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Babes L, Shim R, Kubes P. Imaging α-GalCer-activated iNKT cells in a hepatic metastatic environment. Cancer Immunol Res 2021; 10:12-25. [PMID: 34785505 DOI: 10.1158/2326-6066.cir-21-0445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/12/2021] [Accepted: 11/15/2021] [Indexed: 01/10/2023]
Abstract
Colorectal cancer patients frequently develop liver metastases after, and perhaps as a consequence of, lifesaving surgical resection of the primary tumor. This creates a potential opportunity for prophylactic metastatic treatment with novel immunostimulatory molecules. Here, we used state-of-the-art intravital imaging of an experimental liver metastasis model to visualize the early behavior and function of invariant (i)NKT cells stimulated with α-galactosylceramide (α-GalCer). Intravenous α-GalCer prior to tumor cell seeding in the liver significantly inhibited tumor growth. However, some seeding tumor cells survived. A multiple dosing regimen reduced tumor burden and prolonged the life of mice, whereas tumors returned within 5 days after a single dose of α-GalCer. With multiple doses of α-GalCer, iNKT cells increased in number and granularity (as did NK cells). As a result, the total number of contacts and time in contact with tumors increased substantially. In the absence of iNKT cells, the beneficial effect of α-GalCer was lost. Robust cytokine production dissipated over time. Repeated therapy, even after cytokine dissipation, led to reduced tumor burden and prolonged survival. Serial transplantation of tumors exposed to α-GalCer-activated iNKT cells did not induce greater resistance, suggesting no obvious epigenetic or genetic immunoediting in tumors exposed to activated iNKT cells. Very few tumor cells expressed CD1d in this model, and as such, adding monomers of CD1d-α-GalCer further reduced tumor growth. The data suggest early and repeated stimulation of iNKT cells with α-GalCer could have direct therapeutic benefit for colorectal cancer patients that develop metastatic liver disease.
Collapse
Affiliation(s)
- Liane Babes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute and Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Raymond Shim
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
19
|
Khurana P, Burudpakdee C, Grupp SA, Beier UH, Barrett DM, Bassiri H. Distinct Bioenergetic Features of Human Invariant Natural Killer T Cells Enable Retained Functions in Nutrient-Deprived States. Front Immunol 2021; 12:700374. [PMID: 34434191 PMCID: PMC8380770 DOI: 10.3389/fimmu.2021.700374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/20/2021] [Indexed: 12/19/2022] Open
Abstract
Invariant natural killer T (iNKT) cells comprise a unique subset of lymphocytes that are primed for activation and possess innate NK-like functional features. Currently, iNKT cell-based immunotherapies remain in early clinical stages, and little is known about the ability of these cells to survive and retain effector functions within the solid tumor microenvironment (TME) long-term. In conventional T cells (TCONV), cellular metabolism is linked to effector functions and their ability to adapt to the nutrient-poor TME. In contrast, the bioenergetic requirements of iNKT cells - particularly those of human iNKT cells - at baseline and upon stimulation are not well understood; neither is how these requirements affect effector functions such as production of cytokines and cytolytic proteins. We find that unlike TCONV, human iNKT cells are not dependent upon glucose or glutamine for these effector functions upon stimulation with anti-CD3 and anti-CD28. Additionally, transcriptional profiling revealed that stimulated human iNKT cells are less glycolytic than TCONV and display higher expression of fatty acid oxidation (FAO) and adenosine monophosphate-activated protein kinase (AMPK) pathway genes. Furthermore, stimulated iNKT cells displayed higher mitochondrial mass and membrane potential relative to TCONV. Real-time Seahorse metabolic flux analysis revealed that stimulated human iNKT cells utilize fatty acids as substrates for oxidation more than stimulated TCONV. Together, our data suggest that human iNKT cells possess different bioenergetic requirements from TCONV and display a more oxidative metabolic program relative to effector TCONV. Importantly, iNKT cell-based immunotherapeutic strategies could co-opt such unique features of iNKT cells to improve their efficacy and longevity of anti-tumor responses.
Collapse
Affiliation(s)
- Priya Khurana
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Chakkapong Burudpakdee
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Stephan A. Grupp
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Cell and Therapy Transplant Section, Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ulf H. Beier
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Nephrology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Janssen Research and Development, Spring House, PA, United States
| | | | - Hamid Bassiri
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
20
|
Rudak PT, Choi J, Parkins KM, Summers KL, Jackson DN, Foster PJ, Skaro AI, Leslie K, McAlister VC, Kuchroo VK, Inoue W, Lantz O, Haeryfar SMM. Chronic stress physically spares but functionally impairs innate-like invariant T cells. Cell Rep 2021; 35:108979. [PMID: 33852855 PMCID: PMC8112805 DOI: 10.1016/j.celrep.2021.108979] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 02/09/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
The deleterious effects of psychological stress on mainstream T lymphocytes are well documented. However, how stress impacts innate-like T cells is unclear. We report that long-term stress surprisingly abrogates both T helper 1 (TH1)- and TH2-type responses orchestrated by invariant natural killer T (iNKT) cells. This is not due to iNKT cell death because these cells are unusually refractory to stress-inflicted apoptosis. Activated iNKT cells in stressed mice exhibit a "split" inflammatory signature and trigger sudden serum interleukin-10 (IL-10), IL-23, and IL-27 spikes. iNKT cell dysregulation is mediated by cell-autonomous glucocorticoid receptor signaling and corrected upon habituation to predictable stressors. Importantly, under stress, iNKT cells fail to potentiate cytotoxicity against lymphoma or to reduce the burden of metastatic melanoma. Finally, stress physically spares mouse mucosa-associated invariant T (MAIT) cells but hinders their TH1-/TH2-type responses. The above findings are corroborated in human peripheral blood and hepatic iNKT/MAIT cell cultures. Our work uncovers a mechanism of stress-induced immunosuppression.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Chronic Disease
- Corticosterone/pharmacology
- Cytotoxicity, Immunologic
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immobilization
- Immunity, Innate
- Interleukin-10/genetics
- Interleukin-10/immunology
- Interleukin-23/genetics
- Interleukin-23/immunology
- Interleukins/genetics
- Interleukins/immunology
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Lymphoma/genetics
- Lymphoma/immunology
- Lymphoma/pathology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Mucosal-Associated Invariant T Cells/drug effects
- Mucosal-Associated Invariant T Cells/immunology
- Mucosal-Associated Invariant T Cells/pathology
- Natural Killer T-Cells/drug effects
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/pathology
- Neoplasm Metastasis
- Oxidopamine/pharmacology
- Signal Transduction
- Stress, Psychological/genetics
- Stress, Psychological/immunology
- Stress, Psychological/pathology
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/pathology
- Th1-Th2 Balance
Collapse
Affiliation(s)
- Patrick T Rudak
- Department of Microbiology and Immunology, Western University, London, ON N6A 5C1, Canada
| | - Joshua Choi
- Department of Microbiology and Immunology, Western University, London, ON N6A 5C1, Canada
| | - Katie M Parkins
- Department of Medical Biophysics, Western University, London, ON N6A 5C1, Canada; Robarts Research Institute, Western University, London, ON N6A 5B7, Canada
| | - Kelly L Summers
- Department of Microbiology and Immunology, Western University, London, ON N6A 5C1, Canada
| | - Dwayne N Jackson
- Department of Medical Biophysics, Western University, London, ON N6A 5C1, Canada
| | - Paula J Foster
- Department of Medical Biophysics, Western University, London, ON N6A 5C1, Canada; Robarts Research Institute, Western University, London, ON N6A 5B7, Canada
| | - Anton I Skaro
- Department of Surgery, Division of General Surgery, Western University, London, ON N6A 4V2, Canada
| | - Ken Leslie
- Department of Surgery, Division of General Surgery, Western University, London, ON N6A 4V2, Canada
| | - Vivian C McAlister
- Department of Surgery, Division of General Surgery, Western University, London, ON N6A 4V2, Canada
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Wataru Inoue
- Robarts Research Institute, Western University, London, ON N6A 5B7, Canada; Department of Physiology and Pharmacology, Western University, London, ON N6A 5C1, Canada
| | - Olivier Lantz
- Laboratoire d'Immunologie and INSERM U932, PSL University, Institut Curie, 75248 Paris Cedex 5, France
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, ON N6A 5C1, Canada; Department of Surgery, Division of General Surgery, Western University, London, ON N6A 4V2, Canada; Department of Medicine, Division of Clinical Immunology and Allergy, Western University, London, ON N6A 5A5, Canada.
| |
Collapse
|
21
|
Increased lipid metabolism impairs NK cell function and mediates adaptation to the lymphoma environment. Blood 2021; 136:3004-3017. [PMID: 32818230 DOI: 10.1182/blood.2020005602] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022] Open
Abstract
Natural killer (NK) cells play critical roles in protection against hematological malignancies but can acquire a dysfunctional state, which limits antitumor immunity. However, the underlying reasons for this impaired NK cell function remain to be uncovered. We found that NK cells in aggressive B-cell lymphoma underwent substantial transcriptional reprogramming associated with increased lipid metabolism, including elevated expression of the transcriptional regulator peroxisome activator receptor-γ (PPAR-γ). Exposure to fatty acids in the lymphoma environment potently suppressed NK cell effector response and cellular metabolism. NK cells from both diffuse large B-cell lymphoma patients and Eµ-myc B-cell lymphoma-bearing mice displayed reduced interferon-γ (IFN-γ) production. Activation of PPAR-γ partially restored mitochondrial membrane potential and IFN-γ production. Overall, our data indicate that increased lipid metabolism, while impairing their function, is a functional adaptation of NK cells to the fatty-acid rich lymphoma environment.
Collapse
|
22
|
Rudak PT, Haeryfar SMM. In Vivo Cytotoxicity by α-GalCer-transactivated NK Cells. Methods Mol Biol 2021; 2388:157-174. [PMID: 34524671 DOI: 10.1007/978-1-0716-1775-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Invariant natural killer T (iNKT) cells are innate-like, lipid-reactive T lymphocytes known for their potent immunomodulatory properties. In addition to expressing and utilizing cytolytic effector molecules of their own against certain target cells, iNKT cells can be stimulated with α-galactosylceramide (α-GalCer) to augment the cytotoxic capacity of natural killer (NK) cells. Herein, we describe a flow cytometry-based in vivo killing assay that enables examination of α-GalCer-promoted cytotoxicity against β2 microglobulin knockout (β2M-/-) target cells, which mimic tumor and virus-infected cells displaying little to no MHC class I molecules on their surface. Using an anti-asialo GM1 antibody, which depletes NK cells but not iNKT cells, we confirmed that the increased clearance of β2M-/- cells in α-GalCer-primed recipients was mediated by NK cells. The protocol detailed here can be leveraged to assess the functional fitness of iNKT cells and their crosstalk with NK cells and to further our understanding of α-GalCer-promoted cytotoxicity in preclinical immunotherapeutic applications.
Collapse
Affiliation(s)
- Patrick T Rudak
- Department of Microbiology and Immunology, Western University, London, ON, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, ON, Canada. .,Department of Medicine, Division of Clinical Immunology and Allergy, Western University, London, ON, Canada. .,Department of Surgery, Division of General Surgery, Western University, London, ON, Canada. .,Centre for Human Immunology, Western University, London, ON, Canada. .,Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
23
|
Painter GF, Burn OK, Hermans IF. Using agonists for iNKT cells in cancer therapy. Mol Immunol 2020; 130:1-6. [PMID: 33340930 DOI: 10.1016/j.molimm.2020.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/02/2020] [Indexed: 01/03/2023]
Abstract
The capacity of α-galactosylceramide (α-GalCer) to act as an anti-cancer agent in mice through the specific stimulation of type I NKT (iNKT) cells has prompted extensive investigation to translate this finding to the clinic. However, low frequencies of iNKT cells in cancer patients and their hypo-responsiveness to repeated stimulation have been seen as barriers to its efficacy. Currently the most promising clinical application of α-GalCer, or its derivatives, is as stimuli for ex vivo expansion of iNKT cells for adoptive therapy, although some encouraging clinical results have recently been reported using α-GalCer pulsed onto large numbers of antigen presenting cells (APCs). In on-going preclinical studies, attempts to improve efficacy of injected iNKT cell agonists have focussed on optimising presentation in vivo, through encapsulation in particulate vectors, making structural changes that help binding to the presenting molecule CD1d, or injecting agonists covalently attached to recombinant CD1d. Variations on these same approaches are being used to enhance the APC-licencing function of iNKT cells in vivo to induce adaptive immune responses to associated tumour antigens. Looking ahead, a unique capacity of in vivo-activated iNKT cells to facilitate formation of resident memory CD8+ T cells is a new observation that could find a role in cancer therapy.
Collapse
Affiliation(s)
- Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| | - Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Ian F Hermans
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand; Malaghan Institute of Medical Research, Wellington, New Zealand.
| |
Collapse
|
24
|
Winkler I, Woś J, Bojarska-Junak A, Semczuk A, Rechberger T, Baranowski W, Markut-Miotła E, Tabarkiewicz J, Wolińska E, Skrzypczak M. An association of iNKT+/CD3+/CD161+ lymphocytes in ovarian cancer tissue with CA125 serum concentration. Immunobiology 2020; 225:152010. [PMID: 33130518 DOI: 10.1016/j.imbio.2020.152010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/12/2020] [Accepted: 08/26/2020] [Indexed: 12/01/2022]
Abstract
The purpose of this study was to investigate the association of iNKT (human invariant natural killer T) cells with the key marker of ovarian cancer (OC) - CA125 (cancer antigen125) in serum. The study reports the assessment of iNKT cells in peripheral blood and tissue of benign and borderline ovarian tumors (BOTs) and in the advanced-stage ovarian cancer. The study groups were as follows: 25 women with benign ovarian tumors, 11 women with BOTs, and 24 women with primary advanced-stage ovarian cancers. The control group consisted of 20 patients without the ovarian pathology. The rates of iNKT lymphocytes in the peripheral blood and tissue specimens were evaluated by a flow cytometry. Significant differences in the percentage of iNKT+/CD3+ of CD3+ lymphocytes, iNKT+/CD3+/CD161+ among CD3+ and iNKT+/CD3+/CD161+ among CD3+/iNKT+ between the control group and patients with ovarian tumors in the peripheral blood and tumor tissue were identified. Significant correlations were noticed between the proportion of lymphocytes iNKT+/CD3+/CD161+ among CD3+/iNKT cells in blood and in cancer tissue of both benign and malignant tumors. In the OC group, neither the ratio of iNKT cells in the blood (P = 0.07), nor the intra-tumor NKT-cell infiltration (P = 0.5) were independent prognostic factors for the follow-up. An increased rate of iNKT cells was detected in benign ovarian tumors compared to OCs. In patients with ovarian cancer, a higher rate of iNKT cells in tumor tissue was present related to that noted in the patient's blood. In addition, a correlation was discovered between the CA125 serum marker and NKT cells from the ovarian cancer tissue. This article has for the first time demonstrated a negative relationship between serum levels and NKT lymphocyte count from ovarian tissue. The inflammatory process in ovarian cancer tissue and the potential infiltration of endothelial immune cells, may result in a reduced number of NKT cells in the tumor microenvironment and increased circulation of the CA125 marker. Presented findings underscore new aspects of the iNKT cells involvement in the ovarian cancer development.
Collapse
Affiliation(s)
- Izabela Winkler
- IInd Department of Gynecology, Lublin Medical University, 8 Jaczewski Street, 20-954, Lublin, Poland; IInd Department of Gynecology, St' Johns Center Oncology, 7 Jaczewski Street, 20-090, Lublin, Poland.
| | - Justyna Woś
- Department of Clinical Immunology, Lublin Medical University, 4a Chodźki Street, 20-093, Lublin, Poland
| | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Lublin Medical University, 4a Chodźki Street, 20-093, Lublin, Poland
| | - Andrzej Semczuk
- IInd Department of Gynecology, Lublin Medical University, 8 Jaczewski Street, 20-954, Lublin, Poland
| | - Tomasz Rechberger
- IInd Department of Gynecology, Lublin Medical University, 8 Jaczewski Street, 20-954, Lublin, Poland
| | - Włodzimierz Baranowski
- IInd Department of Gynecology, St' Johns Center Oncology, 7 Jaczewski Street, 20-090, Lublin, Poland; Military Institute of Medicine, Department of Gynecology and Oncological Gynecology, 38 Szaserów street, Warsaw, Poland
| | - Ewa Markut-Miotła
- Department of Pediatric Pulmonology and Rheumatology, Lublin Medical University, 8 Jaczewski Street, 20-090, Lublin, Poland
| | - Jacek Tabarkiewicz
- Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of University of Rzeszów, 1A Warzywna Street, 35-959 Rzeszów, Poland
| | - Ewa Wolińska
- Department of Pathology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Maciej Skrzypczak
- IInd Department of Gynecology, Lublin Medical University, 8 Jaczewski Street, 20-954, Lublin, Poland
| |
Collapse
|
25
|
Enhancing the antitumor functions of invariant natural killer T cells using a soluble CD1d-CD19 fusion protein. Blood Adv 2020; 3:813-824. [PMID: 30858151 DOI: 10.1182/bloodadvances.2018028886] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/17/2019] [Indexed: 12/18/2022] Open
Abstract
Invariant natural killer T (iNKT) cells comprise a unique lineage of CD1d-restricted lipid-reactive T lymphocytes that potently kill tumor cells and exhibit robust immunostimulatory functions. Optimal tumor-directed iNKT cell responses often require expression of the antigen-presenting molecule CD1d on tumors; however, many tumor cells downregulate CD1d and thus evade iNKT cell recognition. We generated a soluble bispecific fusion protein designed to direct iNKT cells to the site of B-cell cancers in a tumor antigen-specific but CD1d-independent manner. This fusion protein is composed of a human CD1d molecule joined to a single chain antibody FV fragment specific for CD19, an antigen widely expressed on B-cell cancers. The CD1d-CD19 fusion protein binds specifically to CD19-expressing, but not CD19-negative cells. Once loaded with the iNKT cell lipid agonist α-galactosyl ceramide (αGC), the CD1d-CD19 fusion induces robust in vitro activation of and cytokine production by human iNKT cells. iNKT cells stimulated by the αGC-loaded CD1d-CD19 fusion also strongly transactivate T-, B-, and NK-cell responses and promote dendritic cell maturation. Importantly, the αGC-loaded fusion induces robust lysis of CD19+CD1d- Epstein-Barr virus immortalized human B-lymphoblastoid cell lines that are otherwise resistant to iNKT cell killing. Consistent with these findings; administration of the αGC-loaded fusion protein controlled the growth of CD19+CD1d- tumors in vivo, suggesting that it can "link" iNKT cells and CD19+CD1d- targets in a therapeutically beneficial manner. Taken together, these preclinical studies demonstrate that this B cell-directed fusion protein can be used to effectively induce iNKT cell antitumor responses in vitro and in vivo.
Collapse
|
26
|
Shimizu K, Iyoda T, Yamasaki S, Kadowaki N, Tojo A, Fujii SI. NK and NKT Cell-Mediated Immune Surveillance against Hematological Malignancies. Cancers (Basel) 2020; 12:cancers12040817. [PMID: 32231116 PMCID: PMC7226455 DOI: 10.3390/cancers12040817] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Recent cancer treatment modalities have been intensively focused on immunotherapy. The success of chimeric antigen receptor T cell therapy for treatment of refractory B cell acute lymphoblastic leukemia has pushed forward research on hematological malignancies. Among the effector types of innate lymphocytes, natural killer (NK) cells show great importance in immune surveillance against infectious and tumor diseases. Particularly, the role of NK cells has been argued in either elimination of target tumor cells or escape of tumor cells from immune surveillance. Therefore, an NK cell activation approach has been explored. Recent findings demonstrate that invariant natural killer T (iNKT) cells capable of producing IFN-γ when optimally activated can promptly trigger NK cells. Here, we review the role of NKT and/or NK cells and their interaction in anti-tumor responses by highlighting how innate immune cells recognize tumors, exert effector functions, and amplify adaptive immune responses. In addition, we discuss these innate lymphocytes in hematological disorders, particularly multiple myeloma and acute myeloid leukemia. The immune balance at different stages of both diseases is explored in light of disease progression. Various types of innate immunity-mediated therapeutic approaches, recent advances in clinical immunotherapies, and iNKT-mediated cancer immunotherapy as next-generation immunotherapy are then discussed.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
- Correspondence: (K.S.); (S.-i.F.); Tel.: +81-45-503-7062 (K.S. & S.-i.F.); Fax: +81-45-503-7061 (K.S. & S.-i.F.)
| | - Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
| | - Norimitsu Kadowaki
- Department of Internal Medicine, Hematology, Rheumatology and Respiratory Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan;
| | - Arinobu Tojo
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Minato, Tokyo 108-8639, Japan;
| | - Shin-ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
- Correspondence: (K.S.); (S.-i.F.); Tel.: +81-45-503-7062 (K.S. & S.-i.F.); Fax: +81-45-503-7061 (K.S. & S.-i.F.)
| |
Collapse
|
27
|
The role of invariant natural killer T cells in experimental xenobiotic-induced cholestatic hepatotoxicity. Biomed Pharmacother 2020; 122:109579. [DOI: 10.1016/j.biopha.2019.109579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/10/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
|
28
|
Fu S, He K, Tian C, Sun H, Zhu C, Bai S, Liu J, Wu Q, Xie D, Yue T, Shen Z, Dai Q, Yu X, Zhu S, Liu G, Zhou R, Duan S, Tian Z, Xu T, Wang H, Bai L. Impaired lipid biosynthesis hinders anti-tumor efficacy of intratumoral iNKT cells. Nat Commun 2020; 11:438. [PMID: 31974378 PMCID: PMC6978340 DOI: 10.1038/s41467-020-14332-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Dysfunction of invariant natural killer T (iNKT) cells in tumor microenvironment hinders their anti-tumor efficacy, and the underlying mechanisms remain unclear. Here we report that iNKT cells increase lipid biosynthesis after activation, and that is promoted by PPARγ and PLZF synergically through enhancing transcription of Srebf1. Among those lipids, cholesterol is required for the optimal IFN-γ production from iNKT cells. Lactic acid in tumor microenvironment reduces expression of PPARγ in intratumoral iNKT cells and consequently diminishes their cholesterol synthesis and IFN-γ production. Importantly, PPARγ agonist pioglitazone, a thiazolidinedione drug for type 2 diabetes, successfully restores IFN-γ production in tumor-infiltrating iNKT cells from both human patients and mouse models. Combination of pioglitazone and alpha-galactosylceramide treatments significantly enhances iNKT cell-mediated anti-tumor immune responses and prolongs survival of tumor-bearing mice. Our studies provide a strategy to augment the anti-tumor efficacy of iNKT cell-based immunotherapies via promoting their lipid biosynthesis. Lipid metabolism has been linked to iNKT function largely as it impacts processing and presentation of lipids they recognize. Here the authors show that iNKT-intrinsic lipid biosynthesis is important for their function but is impaired in tumors, and its restoration with PPARγ agonist drugs promotes anti-tumor iNKT response.
Collapse
Affiliation(s)
- Sicheng Fu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Kaixin He
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Chenxi Tian
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hua Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
| | - Chenwen Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
| | - Shiyu Bai
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Jiwei Liu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Qielan Wu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Di Xie
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ting Yue
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhuxia Shen
- Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Qingqing Dai
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaojun Yu
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shu Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Gang Liu
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230027, China
| | - Rongbin Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Shengzhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Zhigang Tian
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Tao Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
| | - Li Bai
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China. .,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
29
|
Geng J, Wang Y, Zhang L, Wang R, Li C, Sheng W, Li Z, Jiang M. The cajanine derivative LJ101019C regulates the proliferation and enhances the activity of NK cells via Kv1.3 channel-driven activation of the AKT/mTOR pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 66:153113. [PMID: 31790901 DOI: 10.1016/j.phymed.2019.153113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Natural killer (NK) cells play important roles in immune responses and have been wildly used in immunotherapy. Nevertheless, some limitations remain. It is urgent to explore novel and safe strategies to enhance NK cell activity. PURPOSE The aim of this study was to investigate the immuno-stimulatory effects and to reveal the molecular mechanism of LJ101019C, a derivative of a natural small-molecule compounds cajanine, on NK cells. METHODS Cell proliferation was examined by CCK8 assay, then we used the cytotoxicity detection kit to detect the cytotoxicity of NK cells. The change of cell cycle, intracellular reactive oxygen species (ROS) level and mitochondrial mass were evaluated by FACS and Operetta high-content image analysis, respectively. Furthermore, the IFN-γ secretion of NK cells were measured by ELISA. The Kv1.3 protein expression and function were detected by western blot and patch-clamp technique, respectively. The role of Kv1.3 in AKT/mTOR pathway activation was determined by western blot. RESULTS The results showed that LJ101019C at relatively low concentrations (0.05-0.1 µM) significantly increased the proliferation of NK cells. And 1 µM LJ101019C could elevate the proportion of NK cells in the S-phase of the cell cycle (*p < 0.1). Furthermore, the cytotoxic effects of NK cells targeting MIA PaCa-2 cells were significantly enhanced by 0.1 and 1 µM LJ101019C, and were associated with the enhanced secretion of IFN-γ by NK cells (*p < 0.1; **p < 0.05). 0.1 and 1 µM LJ101019C increased intracellular levels of ROS (**p < 0.05), and 0.1 µM LJ101019C elevated mitochondrial mass (*p < 0.1). Electrophysiological recordings indicated that LJ101019C led to a remarkably increase the Kv1.3 current density. Moreover, western blot results indicated that LJ101019C elevated Kv1.3 protein expression and activated AKT/mTOR signaling via increasing the expression of Kv1.3 in NK cells. CONCLUSION LJ101019C increases the proliferation and the cytotoxicity of NK cells at relatively low concentrations. The mechanism is the activation of AKT/mTOR signaling pathway driven by up-regulation of Kv1.3 in NK cells. These suggest LJ101019C is a promising candidate for improving the efficacy of NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Jing Geng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuhong Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lei Zhang
- Department of Organic Chemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ruiqi Wang
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Cong Li
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Weijin Sheng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Zhuorong Li
- Department of Organic Chemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Min Jiang
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
30
|
Bedard M, Shrestha D, Priestman DA, Wang Y, Schneider F, Matute JD, Iyer SS, Gileadi U, Prota G, Kandasamy M, Veerapen N, Besra G, Fritzsche M, Zeissig S, Shevchenko A, Christianson JC, Platt FM, Eggeling C, Blumberg RS, Salio M, Cerundolo V. Sterile activation of invariant natural killer T cells by ER-stressed antigen-presenting cells. Proc Natl Acad Sci U S A 2019; 116:23671-23681. [PMID: 31690657 PMCID: PMC6876220 DOI: 10.1073/pnas.1910097116] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Invariant NKT (iNKT) cells have the unique ability to shape immunity during antitumor immune responses and other forms of sterile and nonsterile inflammation. Recent studies have highlighted a variety of classes of endogenous and pathogen-derived lipid antigens that can trigger iNKT cell activation under sterile and nonsterile conditions. However, the context and mechanisms that drive the presentation of self-lipid antigens in sterile inflammation remain unclear. Here we report that endoplasmic reticulum (ER)-stressed myeloid cells, via signaling events modulated by the protein kinase RNA-like ER kinase (PERK) pathway, increase CD1d-mediated presentation of immunogenic endogenous lipid species, which results in enhanced iNKT cell activation both in vitro and in vivo. In addition, we demonstrate that actin cytoskeletal reorganization during ER stress results in an altered distribution of CD1d on the cell surface, which contributes to enhanced iNKT cell activation. These results define a previously unidentified mechanism that controls iNKT cell activation during sterile inflammation.
Collapse
Affiliation(s)
- Melissa Bedard
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Dilip Shrestha
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - David A Priestman
- Department of Pharmacology, University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Yuting Wang
- Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Falk Schneider
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Juan D Matute
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital Harvard Medical School, Boston, MA 02115
- Division of Neonatology, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Shankar S Iyer
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital Harvard Medical School, Boston, MA 02115
| | - Uzi Gileadi
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Gennaro Prota
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Matheswaran Kandasamy
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Natacha Veerapen
- School of Biosciences, University of Birmingham, B15 2TT Egdbaston, United Kingdom
| | - Gurdyal Besra
- School of Biosciences, University of Birmingham, B15 2TT Egdbaston, United Kingdom
| | - Marco Fritzsche
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
- Kennedy Institute for Rheumatology, University of Oxford, OX3 7LF Oxford, United Kingdom
| | - Sebastian Zeissig
- Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
- Department of Medicine I, University Medical Center Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - John C Christianson
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Science, University of Oxford, OX3 7LD Oxford, United Kingdom
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Christian Eggeling
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
- Institute of Applied Optics and Biophysics, 07743 Jena, Germany
- Department of Biophysical Imaging, Leibniz Institute of Photonic Technologies e.V., 07745 Jena, Germany
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital Harvard Medical School, Boston, MA 02115
| | - Mariolina Salio
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom;
| |
Collapse
|
31
|
Abstract
NKT cells are a small but influential member of the T cell family, recognizing lipids presented by the non-classical MHC-like molecule CD1d rather than peptides presented by classical MHC molecules. They bridge between the innate and adaptive immune systems, serving as rapid responders but also allowing the T cell immune system to recognize lipid antigens, for example derived from tumors or bacteria. They also serve as potent regulatory cells, controlling other immune responses. Type I NKT cells use a semi-invariant T cell receptor (TCR) whereas type II use diverse TCRs. Most often, type I NKT cells promote tumor immunity whereas type II tend to suppress it, and the two subtypes crossregulate each other, forming an immunoregulatory axis. Lack of tools to study these important cells has limited the understanding of these, but newer tools have allowed great advances, especially in mouse models. These range from transgenic and knock-out mice to CD1d tetramers carrying ligands for type I or II NKT cells, to antibodies and NKT cell hybridomas. Here we describe these complementary tools and approaches and their use to study NKT cells and their role in the immunology and immunotherapy of cancer.
Collapse
Affiliation(s)
- Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| | - Purevdorj B Olkhanud
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Masaki Terabe
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
32
|
Wang J, Guillaume J, Janssens J, Remesh SG, Ying G, Bitra A, Van Calenbergh S, Zajonc DM. A molecular switch in mouse CD1d modulates natural killer T cell activation by α-galactosylsphingamides. J Biol Chem 2019; 294:14345-14356. [PMID: 31391251 DOI: 10.1074/jbc.ra119.009963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/31/2019] [Indexed: 11/06/2022] Open
Abstract
Type I natural killer T (NKT) cells are a population of innate like T lymphocytes that rapidly respond to α-GalCer presented by CD1d via the production of both pro- and anti-inflammatory cytokines. While developing novel α-GalCer analogs that were meant to be utilized as potential adjuvants because of their production of pro-inflammatory cytokines (Th1 skewers), we generated α-galactosylsphingamides (αGSA). Surprisingly, αGSAs are not potent antigens in vivo despite their strong T-cell receptor (TCR)-binding affinities. Here, using surface plasmon resonance (SPR), antigen presentation assays, and X-ray crystallography (yielding crystal structures of 19 different binary (CD1d-glycolipid) or ternary (CD1d-glycolipid-TCR) complexes at resolutions between 1.67 and 2.85 Å), we characterized the biochemical and structural details of αGSA recognition by murine NKT cells. We identified a molecular switch within murine (m)CD1d that modulates NKT cell activation by αGSAs. We found that the molecular switch involves a hydrogen bond interaction between Tyr-73 of mCD1d and the amide group oxygen of αGSAs. We further established that the length of the acyl chain controls the positioning of the amide group with respect to the molecular switch and works synergistically with Tyr-73 to control NKT cell activity. In conclusion, our findings reveal important mechanistic insights into the presentation and recognition of glycolipids with polar moieties in an otherwise apolar milieu. These observations may inform the development αGSAs as specific NKT cell antagonists to modulate immune responses.
Collapse
Affiliation(s)
- Jing Wang
- Division of Immune Regulation, La Jolla Institute for Immunology (LJI), La Jolla, California 92037
| | - Joren Guillaume
- Laboratory for Medicinal Chemistry (FFW), Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Jonas Janssens
- Laboratory for Medicinal Chemistry (FFW), Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Soumya G Remesh
- Division of Immune Regulation, La Jolla Institute for Immunology (LJI), La Jolla, California 92037
| | - Ge Ying
- Division of Immune Regulation, La Jolla Institute for Immunology (LJI), La Jolla, California 92037
| | - Aruna Bitra
- Division of Immune Regulation, La Jolla Institute for Immunology (LJI), La Jolla, California 92037
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (FFW), Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Dirk M Zajonc
- Division of Immune Regulation, La Jolla Institute for Immunology (LJI), La Jolla, California 92037 .,Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
33
|
Bröker K, Sinelnikov E, Gustavus D, Schumacher U, Pörtner R, Hoffmeister H, Lüth S, Dammermann W. Mass Production of Highly Active NK Cells for Cancer Immunotherapy in a GMP Conform Perfusion Bioreactor. Front Bioeng Biotechnol 2019; 7:194. [PMID: 31457007 PMCID: PMC6700243 DOI: 10.3389/fbioe.2019.00194] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 07/24/2019] [Indexed: 11/13/2022] Open
Abstract
NK cells have emerged as promising candidates for cancer immunotherapy, especially due to their ability to fight circulating tumor cells thereby preventing metastases formation. Hence several studies have been performed to generate and expand highly cytotoxic NK cells ex vivo, e.g., by using specific cytokines to upregulate both their proliferation and surface expression of distinct activating receptors. Apart from an enhanced activity, application of NK cells as immunotherapeutic agent further requires sufficient cell numbers and a high purity. All these parameters depend on a variety of different factors including the starting material, additives like cytokines as well as the culture system. Here we analyzed PBMC-derived NK cells of five anonymized healthy donors expanded under specific conditions in an innovative perfusion bioreactor system with respect to their phenotype, IFNγ production, and cytotoxicity in vitro. Important features of the meander type bioreactors used here are a directed laminar flow of medium and control of relevant process parameters. Cells are cultivated under "steady state" conditions in perfusion mode. Our data demonstrate that expansion of CD3+ T cell depleted PBMCs in our standardized system generates massive amounts of highly pure (>85%) and potent anti-cancer active NK cells. These cells express a variety of important receptors driving NK cell recruitment, adhesion as well as activation. More specifically, they express the chemokine receptors CXCR3, CXCR4, and CCR7, the adhesion molecules L-selectin, LFA-1, and VLA-4, the activating receptors NKp30, NKp44, NKp46, NKG2D, DNAM1, and CD16 as well as the death ligands TRAIL and Fas-L. Moreover, the generated NK cells show a strong IFNγ expression upon cultivation with K562 tumor cells and demonstrate a high cytotoxicity toward leukemic as well as solid tumor cell lines in vitro. Altogether, these characteristics promise a high clinical potency of thus produced NK cells awaiting further evaluation.
Collapse
Affiliation(s)
- Katharina Bröker
- Center of Internal Medicine II, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany.,Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Udo Schumacher
- Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Pörtner
- Institute of Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | | | - Stefan Lüth
- Center of Internal Medicine II, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany.,Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, The Brandenburg Medical School Theodor Fontane, The University of Potsdam, Potsdam, Germany
| | - Werner Dammermann
- Center of Internal Medicine II, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany.,Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, The Brandenburg Medical School Theodor Fontane, The University of Potsdam, Potsdam, Germany
| |
Collapse
|
34
|
Paul S, Chhatar S, Mishra A, Lal G. Natural killer T cell activation increases iNOS +CD206 - M1 macrophage and controls the growth of solid tumor. J Immunother Cancer 2019; 7:208. [PMID: 31387637 PMCID: PMC6685184 DOI: 10.1186/s40425-019-0697-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/30/2019] [Indexed: 01/12/2023] Open
Abstract
Background NKT cells play an important role in anti-tumor immunity. Alpha-galactosylceramide (α-GalCer), a synthetic glycolipid is presented to natural killer T (NKT) cells by most antigen-presenting cells through CD1d molecules leading to activation of NKT cells. However, the precise mechanisms of how α-GalCer-activated NKT regulate the polarization of the macrophages and effector T cells in the solid tumor are not studied adequately. Methods We induced solid tumor in C57BL/6 mice by subcutaneous injection of B16F10 cell line (1 X 106 cells) and monitored the tumor growth. Animals were given an intraperitoneal injection of α-GalCer (2 μg/injection) in 200 μl PBS on day + 1, + 5, + 10, + 15, and + 20 (with respect to tumor cell injection). Immune cells were characterized using flow cytometry and immunofluorescence staining. NK cells, Gr1+ cells, and F4/80+ macrophages in the mice were depleted by intravenous injection of cell-specific antibodies. Statistical analysis was performed using Student’s t-test or one-way ANOVA. Results Our results showed that intratumoral NKT cells have a lower frequency of CD69, CD25, CD122, and IFN-γR expression; produced less inflammatory cytokines such as IFN-γ, TNF-α, and GM-CSF; higher frequency CD62L+ NKT cells; and also showed reduced proliferation as compared to the splenic NKT cells. Mice treated with α-GalCer showed a significantly increased frequency of IFN-γ-producing NKT cells, CD8+ T cells, and effector Th1 cells. Depletion of NK cells in α-GalCer-treated mice showed a lower frequency of IFN-γ-producing CD4+ and CD8+ T cells in the tumor and prevented the α-GalCer-induced tumor growth. NKT cell activation with α-GalCer treatment significantly increased the iNOS+CD206− M1-macrophages and reduced the iNOS−CD206+ M2-macrophages in the spleen and tumor, and depletion of F4/80+ macrophages prevented the α-GalCer-induced reduction in the tumor growth. Conclusions We showed that activation of NKT cell with α-GalCer modulates the frequency of M1-macrophages and effector Th1 cells in the secondary lymphoid tissues and tumor microenvironment and inhibit tumor growth. The finding suggests that activation of NKT cells with α-GalCer may provide an effective anti-cancer outcome. Electronic supplementary material The online version of this article (10.1186/s40425-019-0697-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sourav Paul
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Sushanta Chhatar
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Amrita Mishra
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Girdhari Lal
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India.
| |
Collapse
|
35
|
Rudak PT, Gangireddy R, Choi J, Burhan AM, Summers KL, Jackson DN, Inoue W, Haeryfar SMM. Stress-elicited glucocorticoid receptor signaling upregulates TIGIT in innate-like invariant T lymphocytes. Brain Behav Immun 2019; 80:793-804. [PMID: 31108170 DOI: 10.1016/j.bbi.2019.05.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/22/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022] Open
Abstract
Stress is known to impede certain host defense mechanisms, including those governed by conventional T lymphocytes. However, whether innate-like T lymphocytes, such as invariant natural killer T (iNKT) and mucosa-associated invariant T (MAIT) cells, are impacted by stress is unclear. Herein, we report that prolonged psychological stress caused by physical confinement results in robust upregulation of T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), an immune checkpoint receptor that controls antitumor and antiviral immune responses. Elevated TIGIT expression was found not only on NK and conventional T cells, but also on iNKT and MAIT cells. Stress-provoked TIGIT upregulation was reversed through treatment with the glucocorticoid receptor (GR) antagonist RU486, but not with 6-hydroxydopamine that induces chemical sympathectomy. A Cre/Lox gene targeting model in which GR was ablated in cells expressing Lck under its proximal promoter revealed that TIGIT upregulation in stressed animals stems from direct GR signaling in T and iNKT cells. In fact, long-term oral administration of exogenous corticosterone (CS) to wild-type C57BL/6 (B6) mice was sufficient to increase TIGIT expression levels on T and iNKT cells. In vitro treatment with CS also potently and selectively upregulated TIGIT, but not CTLA-4 or LAG-3, on mouse iNKT and MAIT hybridomas. These results were recapitulated using primary hepatic iNKT and MAIT cells from wild-type B6 and B6.MAITCAST mice, respectively. Subjecting B6.MAITCAST mice to physical restraint also raised the frequency of TIGIT+ cells among hepatic MAIT cells in a GR-dependent manner. Finally, we found that TIGIT is similarly upregulated in a chronic variable stress model in which animals are exposed to unpredictable heterotypic stressors without developing habituation. Taken together, our findings link, for the first time to our knowledge, GR signaling to TIGIT expression. We propose that glucocorticoid hormones dampen immune responses, in part, by enhancing TIGIT expression across multiple critical subsets of effector lymphocytes, including innate-like T cells. Therefore, TIGIT may constitute an attractive target in immune-enhancing interventions for sustained physiological stress.
Collapse
Affiliation(s)
- Patrick T Rudak
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Rakshith Gangireddy
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Joshua Choi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Amer M Burhan
- Department of Psychiatry, Western University, London, Ontario, Canada
| | - Kelly L Summers
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Dwayne N Jackson
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Wataru Inoue
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Department of Medicine, Division of Clinical Immunology and Allergy, Western University, London, Ontario, Canada; Department of Surgery, Division of General Surgery, Western University, London, Ontario, Canada.
| |
Collapse
|
36
|
Li Z, Wu Y, Wang C, Zhang M. Mouse CD8 +NKT-like cells exert dual cytotoxicity against mouse tumor cells and myeloid-derived suppressor cells. Cancer Immunol Immunother 2019; 68:1303-1315. [PMID: 31278476 PMCID: PMC6682577 DOI: 10.1007/s00262-019-02363-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Our previous work has demonstrated the high efficiency of CD8+ natural killer T (NKT)-like cells in killing antigen-bearing dendritic cells. To evaluate their role in the tumor microenvironment, we performed in vitro and in vivo antitumor experiments to investigate whether CD8+NKT-like cells could kill Yac-1 and B16 cells like NK cells and kill EL4-OVA8 cells in an antigen-specific manner like cytotoxic T lymphocytes (CTLs). Unlike NK1.1−CTLs, CD8+NKT-like cells also exhibit the capability to kill myeloid-derived suppressor cells (MDSCs) in an antigen-specific manner, indicative of their potential role in clearing tumor antigen-bearing MDSCs to improve the antitumor microenvironment. In vitro blocking experiments showed that granzyme B inhibitor efficiently suppressed the cytotoxicity of CD8+NKT-like cells against tumor cells and MDSCs, while Fas ligand (FasL) or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) inhibition failed to produce similar effects. Transcriptomic and phenotypic analyses of CD8+NKT-like cells, NK cells, and NK1.1−CTLs indicated that CD8+NKT-like cells expressed both T-cell activation markers and NK cell markers, thus bearing features of both the activated T cells and NK cells. Taken together, CD8+NKT-like cells could exert NK- and CTL-like antitumor effects through the elimination of both tumor cells and MDSCs in a granzyme B-dependent manner.
Collapse
Affiliation(s)
- Zhengyuan Li
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yiqing Wu
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Chao Wang
- School of Medicine, Tsinghua University, Room B343, Haidian District, Beijing, 100084, China.
| | - Minghui Zhang
- School of Medicine, Tsinghua University, Room B343, Haidian District, Beijing, 100084, China.
| |
Collapse
|
37
|
Zhang Y, Springfield R, Chen S, Li X, Feng X, Moshirian R, Yang R, Yuan W. α-GalCer and iNKT Cell-Based Cancer Immunotherapy: Realizing the Therapeutic Potentials. Front Immunol 2019; 10:1126. [PMID: 31244823 PMCID: PMC6562299 DOI: 10.3389/fimmu.2019.01126] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/03/2019] [Indexed: 01/15/2023] Open
Abstract
NKT cells are CD1d-restricted innate-like T cells expressing both T cell receptor and NK cell markers. The major group of NKT cells in both human and mice is the invariant NKT (iNKT) cells and the best-known function of iNKT cells is their potent anti-tumor function in mice. Since its discovery 25 years ago, the prototype ligand of iNKT cells, α-galactosylceramide (α-GalCer) has been used in over 30 anti-tumor clinical trials with mostly suboptimal outcomes. To realize its therapeutic potential, numerous preclinical models have been developed to optimize the scheme and strategies for α-GalCer-based cancer immunotherapies. Nevertheless, since there is no standard protocol for α-GalCer delivery, we reviewed the preclinical studies with a focus on B16 melanoma model in the goal of identifying the best treatment schemes for α-GalCer treatment. We then reviewed the current progress in developing more clinically relevant mouse models for these preclinical studies, most notably the generation of new mouse models with a humanized CD1d/iNKT cell system. With ever-emerging novel iNKT cell ligands, invention of novel α-GalCer delivery strategies and significantly improved preclinical models for optimizing these new strategies, one can be hopeful that the full potential of anti-tumor potential for α-GalCer will be realized in the not too distant future.
Collapse
Affiliation(s)
- Yingting Zhang
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ryan Springfield
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Siyang Chen
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xiaotian Feng
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rosa Moshirian
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rirong Yang
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
38
|
Nano spray dryer for vectorizing α-galactosylceramide in polymeric nanoparticles: A single step process to enhance invariant Natural Killer T lymphocyte responses. Int J Pharm 2019; 565:123-132. [DOI: 10.1016/j.ijpharm.2019.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/31/2022]
|
39
|
Patel S, Burga RA, Powell AB, Chorvinsky EA, Hoq N, McCormack SE, Van Pelt SN, Hanley PJ, Cruz CRY. Beyond CAR T Cells: Other Cell-Based Immunotherapeutic Strategies Against Cancer. Front Oncol 2019; 9:196. [PMID: 31024832 PMCID: PMC6467966 DOI: 10.3389/fonc.2019.00196] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Chimeric antigen receptor (CAR)-modified T cells have successfully harnessed T cell immunity against malignancies, but they are by no means the only cell therapies in development for cancer. Main Text Summary: Systemic immunity is thought to play a key role in combatting neoplastic disease; in this vein, genetic modifications meant to explore other components of T cell immunity are being evaluated. In addition, other immune cells—from both the innate and adaptive compartments—are in various stages of clinical application. In this review, we focus on these non-CAR T cell immunotherapeutic approaches for malignancy. The first section describes engineering T cells to express non-CAR constructs, and the second section describes other gene-modified cells used to target malignancy. Conclusions: CAR T cell therapies have demonstrated the clinical benefits of harnessing our body's own defenses to combat tumor cells. Similar research is being conducted on lesser known modifications and gene-modified immune cells, which we highlight in this review.
Collapse
Affiliation(s)
- Shabnum Patel
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Rachel A Burga
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Allison B Powell
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Elizabeth A Chorvinsky
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| | - Nia Hoq
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Sarah E McCormack
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Stacey N Van Pelt
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| | - Conrad Russell Y Cruz
- GW Cancer Center, The George Washington University, Washington, DC, United States.,Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| |
Collapse
|
40
|
Amador-Molina A, Trejo-Moreno C, Romero-Rodríguez D, Sada-Ovalle I, Pérez-Cárdenas E, Lamoyi E, Moreno J, Lizano M. Vaccination with human papillomavirus-18 E1 protein plus α-galactosyl-ceramide induces CD8 + cytotoxic response and impairs the growth of E1-expressing tumors. Vaccine 2019; 37:1219-1228. [PMID: 30704821 DOI: 10.1016/j.vaccine.2018.12.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/27/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023]
Abstract
CD8+ T cell-mediated immune response plays a major role in the clearance of virus-infected cells, including human papillomavirus (HPV). The effective treatment of women with normal cytology but persistent high risk-HPV infection or with low-grade intraepithelial lesions could take advantage of novel strategies based on vaccination with viral immunological targets with a wide spectrum of cross-protection. The helicase E1, expressed early during viral replication in HPV infection, is among the most conserved papillomavirus proteins, which makes it a good vaccine candidate. In the present study, we examined E1-specific CD8+ T cell and NK immune responses in a mouse model with α-galactosylceramide (α-GalCer) as an adjuvant. We found that mice immunized with E1 combined with α-GalCer elicited an E1-specific CD8+ T and NK cell cytotoxic responses, which correlated with growth inhibition of grafted melanoma B16-F0 cells expressing E1, both in prophylactic and therapeutic protocols.
Collapse
Affiliation(s)
- Alfredo Amador-Molina
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - Cesar Trejo-Moreno
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - Damaris Romero-Rodríguez
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Tlalpan, Ciudad de México 14080, Mexico
| | - Isabel Sada-Ovalle
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Tlalpan, Ciudad de México 14080, Mexico
| | - Enrique Pérez-Cárdenas
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - Edmundo Lamoyi
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - José Moreno
- Dirección de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico
| | - Marcela Lizano
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, Mexico.
| |
Collapse
|
41
|
Makita Y, Kunii N, Sakurai D, Ihara F, Motohashi S, Suzuki A, Nakayama T, Okamoto Y. Activated iNKT cells enhance the anti-tumor effect of antigen specific CD8 T cells on mesothelin-expressing salivary gland cancer. BMC Cancer 2018; 18:1254. [PMID: 30558663 PMCID: PMC6296067 DOI: 10.1186/s12885-018-5179-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 12/04/2018] [Indexed: 01/22/2023] Open
Abstract
Background Salivary gland cancers are not sensitive to conventional radiotherapy or chemotherapy regimens. Therefore, the development of a new treatment strategy is of critical importance for improving the prognosis. We examined the expression of mesothelin molecules in salivary gland cancers and the efficacy of adoptive cell therapy based on mesothelin-specific chimeric antigen receptor transduced T cells. Methods The expression of mesothelin molecule was studied in salivary gland cancer samples obtained from 16 patients as well as a salivary gland cancer cell line (A-253) and five other cell lines. The activation of mesothelin-specific chimeric antigen receptor-expressing CD8 T cells after stimulation with mesothelin and the effects of invariant natural killer T cells on this activation were evaluated. Results Mesothelin was detected in the A-253 cells and the surgical specimens except for the case of squamous cell carcinoma to various degrees. Following stimulation with mesothelin expressing cancer cells, chimeric antigen receptor T cells were dose-dependently activated; this activation was enhanced by co-culture with invariant natural killer T cells and subsequently abrogated by treatment with anti-interferon-γ antibodies. Furthermore, the cytotoxicity of chimeric antigen receptor T cells against various cancer cells was further augmented by invariant natural killer T cells. Conclusions The use of adoptive transfer with mesothelin-specific chimeric antigen receptor-expressing CD8 T cells against salivary gland cancers is an effective therapy and invariant natural killer T cells are expected to be used in adjuvant treatment for T cell-based immunotherapy. Electronic supplementary material The online version of this article (10.1186/s12885-018-5179-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuji Makita
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Naoki Kunii
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Daiju Sakurai
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Fumie Ihara
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akane Suzuki
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| |
Collapse
|
42
|
Godfrey DI, Le Nours J, Andrews DM, Uldrich AP, Rossjohn J. Unconventional T Cell Targets for Cancer Immunotherapy. Immunity 2018; 48:453-473. [PMID: 29562195 DOI: 10.1016/j.immuni.2018.03.009] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 02/07/2023]
Abstract
Most studies on the immunotherapeutic potential of T cells have focused on CD8 and CD4 T cells that recognize peptide antigens (Ag) presented by polymorphic major histocompatibility complex (MHC) class I and MHC class II molecules, respectively. However, unconventional T cells, which interact with MHC class Ib and MHC-I like molecules, are also implicated in tumor immunity, although their role therein is unclear. These include unconventional T cells targeting MHC class Ib molecules such as HLA-E and its murine ortholog Qa-1b, natural killer T (NKT) cells, mucosal associated invariant T (MAIT) cells, and γδ T cells. Here, we review the current understanding of the roles of these unconventional T cells in tumor immunity and discuss why further studies into the immunotherapeutic potential of these cells is warranted.
Collapse
Affiliation(s)
- Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Jérôme Le Nours
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Daniel M Andrews
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia; Institute of Infection and Immunity, Cardiff University, School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
43
|
Huang W, He W, Shi X, He X, Dou L, Gao Y. The Role of CD1d and MR1 Restricted T Cells in the Liver. Front Immunol 2018; 9:2424. [PMID: 30425710 PMCID: PMC6218621 DOI: 10.3389/fimmu.2018.02424] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/01/2018] [Indexed: 12/17/2022] Open
Abstract
The liver is one of the most important immunological organs that remains tolerogenic in homeostasis yet promotes rapid responses to pathogens in the presence of a systemic infection. The composition of leucocytes in the liver is highly distinct from that of the blood and other lymphoid organs, particularly with respect to enrichment of innate T cells, i.e., invariant NKT cells (iNKT cells) and Mucosal-Associated Invariant T cells (MAIT cells). In recent years, studies have revealed insights into their biology and potential roles in maintaining the immune-environment in the liver. As the primary liver-resident immune cells, they are emerging as significant players in the human immune system and are associated with an increasing number of clinical diseases. As such, innate T cells are promising targets for modifying host defense and inflammation of various liver diseases, including viral, autoimmune, and those of tumor origin. In this review, we emphasize and discuss some of the recent discoveries and advances in the biology of innate T cells, their recruitment and diversity in the liver, and their role in various liver diseases, postulating on their potential application in immunotherapy.
Collapse
Affiliation(s)
- Wenyong Huang
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenjing He
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaomin Shi
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoshun He
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lang Dou
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yifang Gao
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
44
|
Bezgovsek J, Gulbins E, Friedrich SK, Lang KS, Duhan V. Sphingolipids in early viral replication and innate immune activation. Biol Chem 2018; 399:1115-1123. [DOI: 10.1515/hsz-2018-0181] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/21/2018] [Indexed: 01/08/2023]
Abstract
Abstract
In this review, we summarize the mechanisms by which sphingolipids modulate virus multiplication and the host innate immune response, using a number of host-virus systems as illustrative models. Sphingolipids exert diverse functions, both at the level of the viral life cycle and in the regulation of antiviral immune responses. Sphingolipids may influence viral replication in three ways: by serving as (co)receptors during viral entry, by modulating virus replication, and by shaping the antiviral immune response. Several studies have demonstrated that sphingosine kinases (SphK) and their product, sphingosine-1-phosphate (S1P), enhance the replication of influenza, measles, and hepatitis B virus (HBV). In contrast, ceramides, particularly S1P and SphK1, influence the expression of type I interferon (IFN-I) by modulating upstream antiviral signaling and enhancing dendritic cell maturation, differentiation, and positioning in tissue. The synthetic molecule α-galactosylceramide has also been shown to stimulate natural killer cell activation and interferon (IFN)-γ secretion. However, to date, clinical trials have failed to demonstrate any clinical benefit for sphingolipids in the treatment of cancer or HBV infection. Taken together, these findings show that sphingolipids play an important and underappreciated role in the control of virus replication and the innate immune response.
Collapse
|
45
|
Terabe M, Berzofsky JA. Tissue-Specific Roles of NKT Cells in Tumor Immunity. Front Immunol 2018; 9:1838. [PMID: 30158927 PMCID: PMC6104122 DOI: 10.3389/fimmu.2018.01838] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/25/2018] [Indexed: 01/07/2023] Open
Abstract
NKT cells are an unusual population of T cells recognizing lipids presented by CD1d, a non-classical class-I-like molecule, rather than peptides presented by conventional MHC molecules. Type I NKT cells use a semi-invariant T cell receptor and almost all recognize a common prototype lipid, α-galactosylceramide (α-GalCer). Type II NKT cells are any lipid-specific CD1d-restricted T cells that use other receptors and generally don't recognize α-GalCer. They play important regulatory roles in immunity, including tumor immunity. In contrast to type I NKT cells that most have found to promote antitumor immunity, type II NKT cells suppress tumor immunity and the two subsets cross-regulate each other, forming an immunoregulatory axis. They also can promote other regulatory cells including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), and can induce MDSCs to secrete TGF-β, one of the most immunosuppressive cytokines known. In some tumors, both Tregs and type II NKT cells can suppress immunosurveillance, and the balance between these is determined by a type I NKT cell. We have also seen that regulation of tumor immunity can depend on the tissue microenvironment, so the same tumor in the same animal in different tissues may be regulated by different cells, such as type II NKT cells in the lung vs Tregs in the skin. Also, the effector T cells that protect those sites when Tregs are removed do not always act between tissues even in the same animal. Thus, metastases may require different immunotherapy from primary tumors. Newly improved sulfatide-CD1d tetramers are starting to allow better characterization of the elusive type II NKT cells to better understand their function and control it to overcome immunosuppression.
Collapse
Affiliation(s)
- Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
46
|
Sartorius R, D'Apice L, Barba P, Cipria D, Grauso L, Cutignano A, De Berardinis P. Vectorized Delivery of Alpha-GalactosylCeramide and Tumor Antigen on Filamentous Bacteriophage fd Induces Protective Immunity by Enhancing Tumor-Specific T Cell Response. Front Immunol 2018; 9:1496. [PMID: 30002659 PMCID: PMC6031736 DOI: 10.3389/fimmu.2018.01496] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/15/2018] [Indexed: 01/08/2023] Open
Abstract
We have exploited the properties of filamentous bacteriophage fd to deliver immunologically active lipids together with antigenic peptides. Filamentous bacteriophages resemble for size, capability to be permeable to blood vessels, and high density antigen expression, a nature-made nanoparticle. In addition, their major coat protein pVIII, which is arranged to form a tubular shield surrounding the phage genome, has a high content of hydrophobic residues promoting lipid association. We conjugated bacteriophages to alpha-GalactosylCeramide (α-GalCer), a lipid antigen-stimulating invariant natural killer T (iNKT) cells and capable of inducing their anti-tumoral activities. We found that bacteriophage fd/α-GalCer conjugates could repeatedly stimulate iNKT cells in vitro and in vivo, without inducing iNKT anergy. Moreover, co-delivery of α-GalCer and a MHC class I restricted tumor-associated antigenic determinant to antigen-presenting cells via bacteriophages strongly boosted adaptive CD8+ T cell response and efficiently delayed tumor progression. Co-delivery of a tumor antigen and iNKT-stimulatory lipid on the surface of filamentous bacteriophages is a novel approach to potentiate adaptive anti-cancer immune responses, overcoming the current limitations in the use of free α-GalCer and may represent an attractive alternative to existing delivery methods, opening the path to a potential translational usage of this safe, inexpensive, and versatile tool.
Collapse
Affiliation(s)
| | | | - Pasquale Barba
- Institute of Genetics and Biophysics "A. Buzzati Traverso", Naples, Italy
| | | | - Laura Grauso
- Institute of Biomolecular Chemistry (ICB), CNR, Pozzuoli, Italy
| | - Adele Cutignano
- Institute of Biomolecular Chemistry (ICB), CNR, Pozzuoli, Italy
| | | |
Collapse
|
47
|
Rudak PT, Choi J, Haeryfar SMM. MAIT cell-mediated cytotoxicity: Roles in host defense and therapeutic potentials in infectious diseases and cancer. J Leukoc Biol 2018; 104:473-486. [PMID: 29668066 DOI: 10.1002/jlb.4ri0118-023r] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 01/15/2023] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are unconventional, innate-like T lymphocytes that sense the presence of MHC-related protein 1 (MR1)-restricted ligands and select inflammatory cues. Consequently, they release potent immunomodulatory mediators, including IFN-γ, TNF-α, and/or IL-17. MAIT cells can also be viewed as killer cells. They display several NK cell-associated receptors, carry granules containing cytotoxic effector molecules, and swiftly upregulate perforin and granzymes upon activation. Accordingly, MAIT cells are capable of lysing MR1-expressing cells infected with a variety of pathogenic bacteria in in vitro settings and may also mount cytotoxic responses during microbial infections in vivo. Of note, MAIT cell hyperactivation during certain infections may impede their ability to elicit inflammatory and/or cytotoxic responses to secondary stimuli. In addition, MAIT cells isolated from within and from the margin of tumor masses exhibit diminished functions. We propose that MAIT cell-mediated cytotoxicity can be induced, bolstered, or restored to assist in clearing infections and potentially in reducing tumor loads. In this review, we discuss our current understanding of MAIT cells' lytic functions and highlight the pressing questions that need to be addressed in future investigations. We also offer a picture, however hypothetical at this point, of how harnessing the full cytotoxic potentials of MAIT cells may be a valuable approach in the immunotherapy of infectious and malignant diseases.
Collapse
Affiliation(s)
- Patrick T Rudak
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Joshua Choi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Ontario, Canada
| |
Collapse
|
48
|
Keirsse J, Van Damme H, Geeraerts X, Beschin A, Raes G, Van Ginderachter JA. The role of hepatic macrophages in liver metastasis. Cell Immunol 2018; 330:202-215. [PMID: 29661474 DOI: 10.1016/j.cellimm.2018.03.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 03/12/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022]
Abstract
The liver is a major target organ for metastasis of both gastrointestinal and extra-gastrointestinal cancers. Due to its frequently inoperable nature, liver metastasis represents a leading cause of cancer-associated death worldwide. In the past years, the pivotal role of the immune system in this process is being increasingly recognised. In particular, the role of the hepatic macrophages, both recruited monocyte-derived macrophages (Mo-Mfs) and tissue-resident Kupffer cells (KCs), has been shown to be more versatile than initially imagined. However, the lack of tools to easily distinguish between these two macrophage populations has hampered the assignment of particular functionalities to specific hepatic macrophage subsets. In this Review, we highlight the most remarkable findings regarding the origin and functions of hepatic macrophage populations, and we provide a detailed description of their distinct roles in the different phases of the liver metastatic process.
Collapse
Affiliation(s)
- Jiri Keirsse
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Helena Van Damme
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Xenia Geeraerts
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Alain Beschin
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Raes
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
49
|
Wolf BJ, Choi JE, Exley MA. Novel Approaches to Exploiting Invariant NKT Cells in Cancer Immunotherapy. Front Immunol 2018; 9:384. [PMID: 29559971 PMCID: PMC5845557 DOI: 10.3389/fimmu.2018.00384] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/12/2018] [Indexed: 12/24/2022] Open
Abstract
iNKT cells are a subset of innate-like T cells that utilize an invariant TCR alpha chain complexed with a limited repertoire of TCR beta chains to recognize specific lipid antigens presented by CD1d molecules. Because iNKT cells have an invariant TCR, they can be easily identified and targeted in both humans and mice via standard reagents, making this a population of T cells that has been well characterized. iNKT cells are some of the first cells to respond during an infection. By making different types of cytokines in response to different infection stimuli, iNKT cells help determine what kind of immune response then develops. It has been shown that iNKT cells are some of the first cells to respond during infection with a pathogen and the type of cytokines that iNKT cells make help determine the type of immune response that develops in various situations. Indeed, along with immunity to pathogens, pre-clinical mouse studies have clearly demonstrated that iNKT cells play a critical role in tumor immunosurveillance. They can mediate anti-tumor immunity by direct recognition of tumor cells that express CD1d, and/or via targeting CD1d found on cells within the tumor microenvironment. Multiple groups are now working on manipulating iNKT cells for clinical benefit within the context of cancer and have demonstrated that targeting iNKT cells can have a therapeutic benefit in patients. In this review, we briefly introduce iNKT cells, then discuss preclinical data on roles of iNKT cells and clinical trials that have targeted iNKT cells in cancer patients. We finally discuss how future trials could be modified to further increase the efficacy of iNKT cell therapies, in particular CAR-iNKT and rTCR-iNKT cells.
Collapse
Affiliation(s)
| | - Jiyoung Elizabeth Choi
- Agenus Inc., Lexington, MA, United States.,Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Mark A Exley
- Agenus Inc., Lexington, MA, United States.,Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States.,University of Manchester, Manchester, United Kingdom
| |
Collapse
|
50
|
Krijgsman D, Hokland M, Kuppen PJK. The Role of Natural Killer T Cells in Cancer-A Phenotypical and Functional Approach. Front Immunol 2018. [PMID: 29535734 PMCID: PMC5835336 DOI: 10.3389/fimmu.2018.00367] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Natural killer T (NKT) cells are a subset of CD1d-restricted T cells at the interface between the innate and adaptive immune system. NKT cells can be subdivided into functional subsets that respond rapidly to a wide variety of glycolipids and stress-related proteins using T- or natural killer (NK) cell-like effector mechanisms. Because of their major modulating effects on immune responses via secretion of cytokines, NKT cells are also considered important players in tumor immunosurveillance. During early tumor development, T helper (TH)1-like NKT cell subsets have the potential to rapidly stimulate tumor-specific T cells and effector NK cells that can eliminate tumor cells. In case of tumor progression, NKT cells may become overstimulated and anergic leading to deletion of a part of the NKT cell population in patients via activation-induced cell death. In addition, the remaining NKT cells become hyporesponsive, or switch to immunosuppressive TH2-/T regulatory-like NKT cell subsets, thereby facilitating tumor progression and immune escape. In this review, we discuss this important role of NKT cells in tumor development and we conclude that there should be three important focuses of future research in cancer patients in relation with NKT cells: (1) expansion of the NKT cell population, (2) prevention and breaking of NKT cell anergy, and (3) skewing of NKT cells toward TH1-like subsets with antitumor activity.
Collapse
Affiliation(s)
- Daniëlle Krijgsman
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|