1
|
Pinco P, Facciotti F. Unconventional T Cells' Role in Cancer: Unlocking Their Hidden Potential to Guide Tumor Immunity and Therapy. Cells 2025; 14:720. [PMID: 40422223 DOI: 10.3390/cells14100720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/24/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
Unconventional T (UC T) cells, including invariant natural killer T (iNKT) cells, mucosal-associated invariant T (MAIT) cells, γδ T cells, and double-negative (DN) T cells, are key players in immune surveillance and response due to their properties combining innate-like and adaptive-like features. These cells are widely present in mucosal tissues, where they can rapidly respond to infections and tumor-associated changes. In fact, UC T cells can have both pro- and anti-tumoral effects, with their activity influenced by factors such as microbial composition and the tumor microenvironment. In particular, intratumoral microbiota significantly impacts the development, function, and activation of UC T cells, influencing cytokine production and shaping the immune response in various cancers. The complex crosstalk between UC T cells and the surrounding factors is discussed in this review, with a focus on how these cells might be interesting candidates to explore and exploit as anticancer therapeutic agents. However, the great potential of UC T cells, not only demonstrated in the context of adoptive cell transfer, but also enhanced through techniques of engineering, is still flanked by different challenges, like the immunosuppressive tumor microenvironment and heterogeneity of target molecules associated with some specific categories of tumors, like gastrointestinal cancers.
Collapse
Affiliation(s)
- Paola Pinco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Federica Facciotti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
2
|
Singh M, Louie RHY, Samir J, Field MA, Milthorpe C, Adikari T, Mackie J, Roper E, Faulks M, Jackson KJL, Calcino A, Hardy MY, Blombery P, Amos TG, Deveson IW, Wende HV, Floor SN, Read SA, Shek D, Guerin A, Ma CS, Tangye SG, Di Sabatino A, Lenti MV, Pasini A, Ciccocioppo R, Ahlenstiel G, Suan D, Tye-Din JA, Goodnow CC, Luciani F. Expanded T cell clones with lymphoma driver somatic mutations accumulate in refractory celiac disease. Sci Transl Med 2025; 17:eadp6812. [PMID: 40367192 DOI: 10.1126/scitranslmed.adp6812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 03/31/2025] [Indexed: 05/16/2025]
Abstract
Intestinal inflammation continues in a subset of patients with celiac disease despite a gluten-free diet. Here, by applying multi-omic single-cell analysis to duodenal biopsies, we found that low-grade malignancies with lymphoma driver mutations in patients with refractory celiac disease type 2 (RCD2) are comprised by surface CD3-negative (sCD3-) lymphocytes stalled at an innate lymphoid cell (ILC)-progenitor T cell stage undergoing extensive TRA, TRB, and TRD TCR recombination. In people with refractory celiac disease type 1 (RCD1), a disease currently lacking explanation, we identified sCD3+ T cells with lymphoma driver mutations in 6 of 10 individuals with RCD1 and in one of the patients with active, recently diagnosed celiac disease. Furthermore, the mutant T cells formed large TCRαβ clones and displayed inflammatory and cytotoxic molecular profiles. Thus, accumulation of lymphoma driver-mutated T cells and sCD3- progenitors may contribute to chronic, nonresponsive celiac disease.
Collapse
Affiliation(s)
- Mandeep Singh
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Raymond H Y Louie
- School of Computer Science and Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jerome Samir
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Matthew A Field
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Smithfield, QLD 4878, Australia
| | - Claire Milthorpe
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Thiruni Adikari
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Joseph Mackie
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Ellise Roper
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Megan Faulks
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | | | - Andrew Calcino
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Smithfield, QLD 4878, Australia
| | - Melinda Y Hardy
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Piers Blombery
- Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, VIC 3000, Australia
- University of Melbourne, Melbourne, VIC 3010, Australia
| | - Timothy G Amos
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Ira W Deveson
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Helen Vander Wende
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephen N Floor
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Scott A Read
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Dmitri Shek
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Antoine Guerin
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Marco V Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Alessandra Pasini
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona 37134, Italy
| | - Golo Ahlenstiel
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Dan Suan
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jason A Tye-Din
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Gastroenterology Department, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Christopher C Goodnow
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Cellular Genomics Futures Institute and School of Biomedical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Fabio Luciani
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
3
|
Jayasinghe RG, Hollingsworth D, Schedler NC, Landy E, Boonchalermvichian C, Gupta B, Yan H, Baker J, Dejene B, Weinberg KI, Negrin RS, Mavers M. Single-cell transcriptomic profiling reveals diversity in human iNKT cells across hematologic tissues. Cell Rep 2025; 44:115587. [PMID: 40305288 DOI: 10.1016/j.celrep.2025.115587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/20/2024] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Invariant natural killer T (iNKT) cells are evolutionarily conserved innate lymphocytes important for protection against pathogens, malignancies, and graft-versus-host disease, with potential for universal donor cellular therapies. While mouse studies reveal transcriptionally and functionally distinct subsets, a comprehensive understanding of human iNKT cell heterogeneity is limited. Herein, we delineate the transcriptomic diversity of human iNKT cells from multiple immunologically relevant hematologic tissues. Human iNKT cells express naive/precursor, transitional, and T helper (Th)1/17/NK-like transcriptional profiles, partially contrasting with findings in mice. Additionally, these data uncover transcription factor dynamics not previously described in mice and reveal a T effector memory RA+-like population. Further, two distinct expression patterns of human CD8+ iNKT cells are described-one resembling naive/precursor cells and another resembling Th1/17/NK-like cells, with predominant expression of CD8αα protein. These critical insights into the transcriptional heterogeneity of human iNKT cells will facilitate future functional studies and inform iNKT-based cellular therapy development.
Collapse
Affiliation(s)
- Reyka G Jayasinghe
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Derek Hollingsworth
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathan C Schedler
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Emily Landy
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chaiyaporn Boonchalermvichian
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Biki Gupta
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Hao Yan
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeanette Baker
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Beruh Dejene
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kenneth I Weinberg
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert S Negrin
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Melissa Mavers
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
4
|
Hosono Y, Tomiyasu N, Kasai H, Ishikawa E, Takahashi M, Imamura A, Ishida H, Compostella F, Kida H, Kumanogoh A, Bamba T, Izumi Y, Yamasaki S. Identification of α-galactosylceramide as an endogenous mammalian antigen for iNKT cells. J Exp Med 2025; 222:e20240728. [PMID: 39704712 DOI: 10.1084/jem.20240728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/08/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
Invariant natural killer T (iNKT) cells are unconventional T cells recognizing lipid antigens in a CD1d-restricted manner. Among these lipid antigens, α-galactosylceramide (α-GalCer), which was originally identified in marine sponges, is the most potent antigen. Although the presence of α-anomeric hexosylceramide and microbiota-derived branched α-GalCer is reported, antigenic α-GalCer has not been identified in mammals. Here, we developed a high-resolution separation and detection system, supercritical fluid chromatography tandem mass spectrometry (SFC/MS/MS), that can discriminate hexosylceramide diastereomers (α-GalCer, α-GlcCer, β-GalCer, or β-GlcCer). The B16 melanoma tumor cell line does not activate iNKT cells; however, ectopic expression of CD1d was sufficient to activate iNKT cells without adding antigens. B16 melanoma was unlikely to generate iNKT cell antigens; instead, antigen activity was detected in cell culture serum. Activity-based purification and SFC/MS/MS identified dihydrosphingosine-based saturated α-GalCer as an antigenic component in serum, bile, and lymphoid tissues. These results show the first evidence for the presence of potent antigenic α-GalCer in mammals.
Collapse
Affiliation(s)
- Yuki Hosono
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University , Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyuki Tomiyasu
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Hayato Kasai
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University , Suita, Japan
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University , Suita, Japan
| | - Masatomo Takahashi
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Akihiro Imamura
- Department of Applied Bioorganic Chemistry, Gifu University, Gifu, Japan
- Institute for Glyco-core Research, Gifu University , Gifu, Japan
| | - Hideharu Ishida
- Department of Applied Bioorganic Chemistry, Gifu University, Gifu, Japan
- Institute for Glyco-core Research, Gifu University , Gifu, Japan
| | - Federica Compostella
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Hiroshi Kida
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Immunopathology, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University , Suita, Japan
- Center for Advanced Modalities and DDS, Osaka University , Suita, Japan
| | - Takeshi Bamba
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Izumi
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University , Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University , Suita, Japan
- Center for Advanced Modalities and DDS, Osaka University , Suita, Japan
| |
Collapse
|
5
|
Lagattuta KA, Kohlgruber AC, Abdelfattah NS, Nathan A, Rumker L, Birnbaum ME, Elledge SJ, Raychaudhuri S. The T cell receptor sequence influences the likelihood of T cell memory formation. Cell Rep 2025; 44:115098. [PMID: 39731734 PMCID: PMC11785489 DOI: 10.1016/j.celrep.2024.115098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/19/2024] [Accepted: 12/02/2024] [Indexed: 12/30/2024] Open
Abstract
The amino acid sequence of the T cell receptor (TCR) varies between T cells of an individual's immune system. Particular TCR residues nearly guarantee mucosal-associated invariant T (MAIT) and natural killer T (NKT) cell transcriptional fates. To define how the TCR sequence affects T cell fates, we analyze the paired αβTCR sequence and transcriptome of 961,531 single cells. We find that hydrophobic complementarity-determining region (CDR)3 residues promote regulatory T cell fates in both the CD8 and CD4 lineages. Most strikingly, we find a set of TCR sequence features that promote the T cell transition from naive to memory. We quantify the extent of these features through our TCR scoring function "TCR-mem." Using TCR transduction experiments, we demonstrate that increased TCR-mem promotes T cell activation, even among T cells that recognize the same antigen. Our results reveal a common set of TCR sequence features that enable T cell activation and immunological memory.
Collapse
MESH Headings
- Immunologic Memory/immunology
- Animals
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Mice
- Memory T Cells/immunology
- Amino Acid Sequence
- Lymphocyte Activation/immunology
- Complementarity Determining Regions/immunology
- Mice, Inbred C57BL
- Receptors, Antigen, T-Cell, alpha-beta
- CD8-Positive T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Kaitlyn A Lagattuta
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ayano C Kohlgruber
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Nouran S Abdelfattah
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Laurie Rumker
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Stephen J Elledge
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Golzari-Sorkheh M, Yoganathan K, Chen ELY, Singh J, Zúñiga-Pflücker JC. T Cell Development: From T-Lineage Specification to Intrathymic Maturation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:81-137. [PMID: 40067585 DOI: 10.1007/978-3-031-77921-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
T cell development occurs in the thymus in both mice and humans. Upon entry into the thymus, bone marrow-derived blood-borne progenitors receive instructive signals, including Notch signaling, to eliminate their potential to develop into alternative immune lineages while committing to the T cell fate. Upon T-lineage commitment, developing T cells receive further instructional cues to generate different T cell sublineages, which together possess diverse immunological functions to provide host immunity. Over the years, numerous studies have contributed to a greater understanding of key thymic signals that govern T cell differentiation and subset generation. Here, we review these critical signaling factors that govern the different stages of both mouse and human T cell development, while also focusing on the transcriptional changes that mediate T cell identity and diversity.
Collapse
Affiliation(s)
- Mahdieh Golzari-Sorkheh
- Department of Immunology, University of Toronto & Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kogulan Yoganathan
- Department of Immunology, University of Toronto & Sunnybrook Research Institute, Toronto, ON, Canada
| | - Edward L Y Chen
- Department of Immunology, University of Toronto & Sunnybrook Research Institute, Toronto, ON, Canada
| | - Jastaranpreet Singh
- Department of Immunology, University of Toronto & Sunnybrook Research Institute, Toronto, ON, Canada
| | | |
Collapse
|
7
|
Nishio K, Hsu KS, Berzofsky JA, Olkhanud PB. NKT Hybridoma Cell Stimulation Assays to Evaluate Glycolipid Specificity and Processing. Methods Mol Biol 2025; 2930:93-102. [PMID: 40402450 DOI: 10.1007/978-1-0716-4558-1_8] [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] [Indexed: 05/23/2025]
Abstract
Natural Killer T (NKT) cells, a specialized subset of T cells, play a pivotal role in immune surveillance. They recognize lipid antigens presented by the MHC class I-like molecule CD1d, bridging innate and adaptive immunity. NKT cells exhibit both pro-inflammatory and immunoregulatory properties, impacting various pathological states, including infections, autoimmune diseases, and cancer. The NKT hybridoma cell stimulation assay is a powerful tool for detecting and characterizing NKT cells, as well as screening natural and synthetic lipid antigens, potentially leading to novel therapeutic interventions. Here, we describe two complementary methods (cell-free and cell-based) to stimulate NKT hybridoma cells to secrete cytokine, which serves as an indirect measure of NKT cell activation. These assays provide insight into the intricate interplay between NKT cells, lipids, and other immune cell subsets, including antigen-presenting cells, and their impact on immune regulation and disease progression.
Collapse
Affiliation(s)
- Kumiko Nishio
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Kevin S Hsu
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Purevdorj B Olkhanud
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
8
|
Wang Z, Zhang G. CAR-iNKT cell therapy: mechanisms, advantages, and challenges. Curr Res Transl Med 2025; 73:103488. [PMID: 39662251 DOI: 10.1016/j.retram.2024.103488] [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: 11/04/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
In recent years, chimeric antigen receptor (CAR) T-cell therapy has emerged as a groundbreaking approach in cancer immunotherapy. Particularly in hematologic malignancies, such as B-cell acute lymphoblastic leukemia (B-ALL), B cell lymphomas and multiple myeloma. CAR-T therapy has demonstrated remarkable clinical efficacy, leading to the approval of several CAR-T cell products and offering significant benefits to numerous leukemia patients. Despite these successes, the application of CAR-T cells in solid tumors remains limited due to significant challenges, including immunosuppressive tumor microenvironments, heterogeneous antigen expression, and treatment-associated toxicities. In parallel with CAR-T development, researchers are investigating other immune cell platforms to overcome these obstacles. Among these, invariant natural killer T (iNKT) cells have garnered increasing attention for their unique immunological properties. Unlike conventional T cells, iNKT cells are a subset of T lymphocytes characterized by the expression of a semi-invariant T-cell receptor (TCR) that recognizes lipid antigens presented by CD1d molecules. This distinctive antigen recognition mechanism enables iNKT cells to bridge innate and adaptive immunity, granting them potent antitumor activity and the ability to modulate the tumor microenvironment. Additionally, iNKT cells exhibit intrinsic resistance to exhaustion and an enhanced ability to infiltrate solid tumors compared to traditional T cells. Building on these properties, researchers are leveraging CAR technology to enhance iNKT cell tumor-targeting capabilities, aiming to overcome barriers encountered in solid tumor therapy. This review provides an in-depth discussion of the application and therapeutic potential of CAR-iNKT cells in cancer immunotherapy, with a focus on their advantages over conventional CAR-T cells and their role in addressing the challenges of solid tumor treatment.
Collapse
Affiliation(s)
- Zixuan Wang
- Beijing Institute of Biological Products Co., Ltd, Beijing 101149, China
| | - Guangji Zhang
- Beijing Rongai Biotechnology Co., Ltd, 1st Floor, Building 29, No. 5 Kechuang East 2nd Street, Tongzhou District, Beijing 101100, China.
| |
Collapse
|
9
|
Morris I, Vrieling F, Bouwman A, Stienstra R, Kalkhoven E. Lipid accumulation in adipose tissue-resident iNKT cells contributes to an inflammatory phenotype. Adipocyte 2024; 13:2421750. [PMID: 39484712 PMCID: PMC11540091 DOI: 10.1080/21623945.2024.2421750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/03/2024] Open
Abstract
Reciprocal communication between adipocytes and immune cells is essential to maintain optimal adipose tissue (AT) functionality. Amongst others, adipocytes directly interact with invariant NKT cells (iNKT cells), which in turn secrete various cytokines. A lipid-rich microenvironment, as observed in obesity, skews this adipocyte-driven cytokine output towards a more inflammatory output. Whether a lipid-rich microenvironment also affects iNKT cells directly, however, is unknown. Here, we show that primary mouse iNKT cells isolated from AT can accumulate lipids in lipid droplets (LDs), more so than liver- and spleen-resident iNKT cells. Furthermore, a lipid-rich microenvironment increased the production of the proinflammatory cytokine IFNγ. Next, to an indirect, adipocyte-mediated cue, iNKT cells can directly respond to environmental lipid changes, supporting a potential role as nutrient sensors.
Collapse
Affiliation(s)
- Imogen Morris
- Ce nter for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Frank Vrieling
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Annemieke Bouwman
- Ce nter for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rinke Stienstra
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eric Kalkhoven
- Ce nter for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
10
|
Shyanti RK, Haque M, Singh R, Mishra M. Optimizing iNKT-driven immune responses against cancer by modulating CD1d in tumor and antigen presenting cells. Clin Immunol 2024; 269:110402. [PMID: 39561929 DOI: 10.1016/j.clim.2024.110402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Two major antigen processing pathways represent protein Ags through major histocompatibility complexes (MHC class I and II) or lipid Ags through CD1 molecules influence the tumor immune response. Invariant Natural Killer T cells (iNKT) manage a significant role in cancer immunotherapy. CD1d, found on antigen-presenting cells (APCs), presents lipid Ags to iNKT cells. In many cancers, the number and function of iNKT cell are compromised, leading to immune evasion. Additionally impaired motility of iNKT cells may contribute to poor tumor prognosis. Emerging evidences suggest that CD1d, itself also influences cancer progression. Patient databases further highlight the importance of CD1d expression in different cancers and its correlation with patient survival outcomes. The ability of iNKT cells to activate and enhance the immune response renders them an attractive target for cancer immunotherapy. This review discusses all the possible ways of cancer immune evasion and restoration of immune responses mediated by CD1d-iNKT interactions.
Collapse
Affiliation(s)
- Ritis Kumar Shyanti
- Cancer Research Center, Department of Biological Sciences, Alabama State University, AL 36104, USA
| | - Mazharul Haque
- Cancer Research Center, Department of Biological Sciences, Alabama State University, AL 36104, USA
| | - Rajesh Singh
- Microbiology, Biochemistry, and Immunology, Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Manoj Mishra
- Cancer Research Center, Department of Biological Sciences, Alabama State University, AL 36104, USA.
| |
Collapse
|
11
|
Derakhshandeh R, Zhu Y, Li J, Hester D, Younis R, Koka R, Jones LP, Sun W, Goloubeva O, Tkaczuk K, Bates J, Reader J, Webb TJ. Identification of Functional Immune Biomarkers in Breast Cancer Patients. Int J Mol Sci 2024; 25:12309. [PMID: 39596374 PMCID: PMC11595306 DOI: 10.3390/ijms252212309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer immunotherapy has emerged as an effective, personalized treatment for certain patients, particularly for those with hematological malignancies. However, its efficacy in breast cancer has been marginal-perhaps due to cold, immune-excluded, or immune-desert tumors. Natural killer T (NKT) cells play a critical role in cancer immune surveillance and are reduced in cancer patients. Thus, we hypothesized that NKT cells could serve as a surrogate marker for immune function. In order to assess which breast cancer patients would likely benefit from immune cell-based therapies, we have developed a quantitative method to rapidly assess NKT function using stimulation with artificial antigen presenting cells followed by quantitative real-time PCR for IFN-γ. We observed a significant reduction in the percentage of circulating NKT cells in breast cancer patients, compared to healthy donors; however, the majority of patients had functional NKT cells. When we compared BC patients with highly functional NKT cells, as indicated by high IFN-γ induction, to those with little to no induction, following stimulation of NKT cells, there was no significant difference in NKT cell number between the groups, suggesting functional loss has more impact than physical loss of this subpopulation of T cells. In addition, we assessed the percentage of tumor-infiltrating lymphocytes and PD-L1 expression within the tumor microenvironment in the low and high responders. Further characterization of immune gene signatures in these groups identified a concomitant decrease in the induction of TNFα, LAG3, and LIGHT in the low responders. We next investigated the mechanisms by which breast cancers suppress NKT-mediated anti-tumor immune responses. We found that breast cancers secrete immunosuppressive lipids, and treatment with commonly prescribed medications that modulate lipid metabolism, can reduce tumor growth and restore NKT cell responses.
Collapse
Affiliation(s)
- Roshanak Derakhshandeh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Yuyi Zhu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Junxin Li
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Danubia Hester
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Rania Younis
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA;
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
| | - Rima Koka
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Laundette P. Jones
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wenji Sun
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Olga Goloubeva
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Katherine Tkaczuk
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
| | - Joshua Bates
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Jocelyn Reader
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tonya J. Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
| |
Collapse
|
12
|
Rajashekar V, Stern L, Almeida CF, Slobedman B, Abendroth A. The surveillance of viral infections by the unconventional Type I NKT cell. Front Immunol 2024; 15:1472854. [PMID: 39355244 PMCID: PMC11442276 DOI: 10.3389/fimmu.2024.1472854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/26/2024] [Indexed: 10/03/2024] Open
Abstract
Type I NKT cells, also known as Invariant Natural Killer T (iNKT) cells, are a subpopulation of unconventional, innate-like T (ILT) cells which can proficiently influence downstream immune effector functions. Type I NKT cells express a semi-invariant αβ T cell receptor (TCR) that recognises lipid-based ligands specifically presented by the non-classical cluster of differentiation (CD1) protein d (CD1d) molecule. Due to their potent immunomodulatory functional capacity, type I NKT cells are being increasingly considered in prophylactic and therapeutic approaches towards various diseases, including as vaccine-adjuvants. As viruses do not encode lipid synthesis, it is surprising that many studies have shown that some viruses can directly impede type I NKT activation through downregulating CD1d expression. Therefore, in order to harness type I NKT cells for potential anti-viral therapeutic uses, it is critical that we fully appreciate how the CD1d-iNKT cell axis interacts with viral immunity. In this review, we examine clinical findings that underpin the importance of type I NKT cell function in viral infections. This review also explores how certain viruses employ immunoevasive mechanisms and directly encode functions to target CD1d expression and type I NKT cell function. Overall, we suggest that the CD1d-iNKT cell axis may hold greater gravity within viral infections than what was previously appreciated.
Collapse
Affiliation(s)
- Varshini Rajashekar
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Lauren Stern
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Catarina F. Almeida
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Barry Slobedman
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Allison Abendroth
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Romanò C, Jiang H, Tahvili S, Wei P, Keiding UB, Clergeaud G, Skovbakke SL, Blomberg AL, Hafkenscheid L, Henriksen JR, Andresen TL, Goletz S, Hansen AE, Christensen D, Clausen MH. Chemical synthesis and immunological evaluation of cancer vaccines based on ganglioside antigens and α-galactosylceramide. RSC Med Chem 2024; 15:2718-2728. [PMID: 39149099 PMCID: PMC11324045 DOI: 10.1039/d4md00387j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/20/2024] [Indexed: 08/17/2024] Open
Abstract
iNKT cells - often referred as the "Swiss Army knife" of the immune system - have emerged as central players in cancer vaccine therapies. Glycolipids activating iNKT cells, such as α-galactosylceramide (αGalCer), can enhance the immune response against co-delivered cancer antigens and have been applied in the design of self-adjuvanting anti-tumor vaccines. In this context, this work focuses on the chemical synthesis of ganglioside tumor-associated carbohydrate antigens (TACAs), namely GM3 and (Neu5Gc)GM3 antigens, their conjugation to αGalCer, and their formulation into liposomes as an efficient platform for their in vivo delivery. Liposomes containing GM3-αGalCer, (Neu5Gc)GM3-αGalCer, and equimolar amounts of the two conjugates have been fully characterized and their ability to activate iNKT cell has been confirmed ex vivo in mouse and human cell assays. The candidates were tested in in vivo immunization studies, demonstrating an ability to induce both TH1 and TH2 cytokines leading to the production of all subclasses of IgG antibodies. Notably, the study also demonstrated that serum antibodies raised against the two TACAs, alone and in combination, were cross-reactive. This finding has consequences for future vaccine designs - even if a highly tumor-selective antigen is chosen, the resulting antibody response may be broader than anticipated.
Collapse
Affiliation(s)
- Cecilia Romanò
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Hao Jiang
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Sahar Tahvili
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Peng Wei
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Ulrik B Keiding
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Gael Clergeaud
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark Ørsteds Plads 2800 Kgs Lyngby Denmark
| | - Sarah Line Skovbakke
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark Søltofts Plads 2800 Kgs Lyngby Denmark
| | - Anne Louise Blomberg
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark Søltofts Plads 2800 Kgs Lyngby Denmark
| | - Lise Hafkenscheid
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark Søltofts Plads 2800 Kgs Lyngby Denmark
| | - Jonas R Henriksen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark Ørsteds Plads 2800 Kgs Lyngby Denmark
| | - Thomas L Andresen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark Ørsteds Plads 2800 Kgs Lyngby Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark Søltofts Plads 2800 Kgs Lyngby Denmark
| | - Anders E Hansen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark Ørsteds Plads 2800 Kgs Lyngby Denmark
| | - Dennis Christensen
- Adjuvant Systems Research & Development, Croda Pharma 2800 Lyngby Denmark
| | - Mads H Clausen
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| |
Collapse
|
14
|
O’Neal J, Mavers M, Jayasinghe RG, DiPersio JF. Traversing the bench to bedside journey for iNKT cell therapies. Front Immunol 2024; 15:1436968. [PMID: 39170618 PMCID: PMC11335525 DOI: 10.3389/fimmu.2024.1436968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Invariant natural killer T (iNKT) cells are immune cells that harness properties of both the innate and adaptive immune system and exert multiple functions critical for the control of various diseases. Prevention of graft-versus-host disease (GVHD) by iNKT cells has been demonstrated in mouse models and in correlative human studies in which high iNKT cell content in the donor graft is associated with reduced GVHD in the setting of allogeneic hematopoietic stem cell transplants. This suggests that approaches to increase the number of iNKT cells in the setting of an allogeneic transplant may reduce GVHD. iNKT cells can also induce cytolysis of tumor cells, and murine experiments demonstrate that activating iNKT cells in vivo or treating mice with ex vivo expanded iNKT cells can reduce tumor burden. More recently, research has focused on testing anti-tumor efficacy of iNKT cells genetically modified to express a chimeric antigen receptor (CAR) protein (CAR-iNKT) cells to enhance iNKT cell tumor killing. Further, several of these approaches are now being tested in clinical trials, with strong safety signals demonstrated, though efficacy remains to be established following these early phase clinical trials. Here we review the progress in the field relating to role of iNKT cells in GVHD prevention and anti- cancer efficacy. Although the iNKT field is progressing at an exciting rate, there is much to learn regarding iNKT cell subset immunophenotype and functional relationships, optimal ex vivo expansion approaches, ideal treatment protocols, need for cytokine support, and rejection risk of iNKT cells in the allogeneic setting.
Collapse
Affiliation(s)
- Julie O’Neal
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, United States
| | - Melissa Mavers
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, United States
- Division of Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Reyka G. Jayasinghe
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
15
|
Gao M, Zhao X. Insights into the tissue repair features of MAIT cells. Front Immunol 2024; 15:1432651. [PMID: 39086492 PMCID: PMC11289772 DOI: 10.3389/fimmu.2024.1432651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are a subset of innate-like non-conventional T cells characterized by multifunctionality. In addition to their well-recognized antimicrobial activity, increasing attention is being drawn towards their roles in tissue homeostasis and repair. However, the precise mechanisms underlying these functions remain incompletely understood and are still subject to ongoing exploration. Currently, it appears that the tissue localization of MAIT cells and the nature of the diseases or stimuli, whether acute or chronic, may induce a dynamic interplay between their pro-inflammatory and anti-inflammatory, or pathogenic and reparative functions. Therefore, elucidating the conditions and mechanisms of MAIT cells' reparative functions is crucial for fully maximizing their protective effects and advancing future MAIT-related therapies. In this review, we will comprehensively discuss the establishment and potential mechanisms of their tissue repair functions as well as the translational application prospects and current challenges in this field.
Collapse
Affiliation(s)
- Mengge Gao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaosu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| |
Collapse
|
16
|
Cruz de Casas P, Knöpper K, Dey Sarkar R, Kastenmüller W. Same yet different - how lymph node heterogeneity affects immune responses. Nat Rev Immunol 2024; 24:358-374. [PMID: 38097778 DOI: 10.1038/s41577-023-00965-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 05/04/2024]
Abstract
Lymph nodes are secondary lymphoid organs in which immune responses of the adaptive immune system are initiated and regulated. Distributed throughout the body and embedded in the lymphatic system, local lymph nodes are continuously informed about the state of the organs owing to a constant drainage of lymph. The tissue-derived lymph carries products of cell metabolism, proteins, carbohydrates, lipids, pathogens and circulating immune cells. Notably, there is a growing body of evidence that individual lymph nodes differ from each other in their capacity to generate immune responses. Here, we review the structure and function of the lymphatic system and then focus on the factors that lead to functional heterogeneity among different lymph nodes. We will discuss how lymph node heterogeneity impacts on cellular and humoral immune responses and the implications for vaccination, tumour development and tumour control by immunotherapy.
Collapse
Affiliation(s)
- Paulina Cruz de Casas
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Konrad Knöpper
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Rupak Dey Sarkar
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Wolfgang Kastenmüller
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
17
|
Xu X, Zhang J, Xing H, Han L, Li X, Wu P, Tang J, Jing L, Luo J, Luo J, Liu L. Identification of metabolism-related key genes as potential biomarkers for pathogenesis of immune thrombocytopenia. Sci Rep 2024; 14:9040. [PMID: 38641637 PMCID: PMC11031595 DOI: 10.1038/s41598-024-59493-7] [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: 01/03/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024] Open
Abstract
Immune thrombocytopenia (ITP), an acquired autoimmune disease, is characterized by immune-mediated platelet destruction. A biomarker is a biological entity that contributes to disease pathogenesis and reflects disease activity. Metabolic alterations are reported to be associated with the occurrence of various diseases. As metabolic biomarkers for ITP have not been identified. This study aimed to identify metabolism-related differentially expressed genes as potential biomarkers for pathogenesis of ITP using bioinformatic analyses.The microarray expression data of the peripheral blood mononuclear cells were downloaded from the Gene Expression Omnibus database (GSE112278 download link: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE112278 ). Key module genes were intersected with metabolism-related genes to obtain the metabolism-related key candidate genes. The hub genes were screened based on the degree function in the coytoscape sofware. The key ITP-related genes were subjected to functional enrichment analysis. Immune infiltration analysis was performed using a single-sample gene set enrichment analysis algorithm to evaluate the differential infiltration levels of immune cell types between ITP patient and control. Molecular subtypes were identified based on the expression of hub genes. The expression of hub genes in the ITP patients was validated using quantitative real-time polymerase chain reaction analysis. This study identified five hub genes (ADH4, CYP7A1, CYP1A2, CYP8B1, and NR1H4), which were be associated with the pathogenesis of ITP, and two molecular subtypes of ITP. Among these hub genes, CYP7A1 and CYP8B1 involved in cholesterol metabolism,were further verified in clinical samples.
Collapse
Affiliation(s)
- Xiangmei Xu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jiamin Zhang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Hongyun Xing
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Liying Han
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Xiaoming Li
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Pengqiang Wu
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jirui Tang
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Li Jing
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jie Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Jing Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
18
|
Moraes Ribeiro E, Secker KA, Nitulescu AM, Schairer R, Keppeler H, Wesle A, Schmid H, Schmitt A, Neuber B, Chmiest D, Podavini S, Märklin M, Klimovich B, Schmitt M, Korkmaz F, Lengerke C, Schneidawind C, Schneidawind D. PD-1 checkpoint inhibition enhances the antilymphoma activity of CD19-CAR-iNKT cells that retain their ability to prevent alloreactivity. J Immunother Cancer 2024; 12:e007829. [PMID: 38296597 PMCID: PMC10831439 DOI: 10.1136/jitc-2023-007829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Relapse and graft-versus-host disease (GVHD) are the main causes of death after allogeneic hematopoietic cell transplantation (HCT). Preclinical murine models and clinical data suggest that invariant natural killer T (iNKT) cells prevent acute and chronic GVHD. In addition, iNKT cells are crucial for efficient immune responses against malignancies and contribute to reduced relapse rates after transplantation. Chimeric antigen receptors (CAR) redirect effector cells to cell surface antigens and enhance killing of target cells. With this study, we aimed to combine enhanced cytotoxicity of CD19-CAR-iNKT cells against lymphoma cells with their tolerogenic properties. METHODS iNKT cells were isolated from peripheral blood mononuclear cells and transduced with an anti-CD19-CAR retrovirus. After in vitro expansion, the functionality of CD19-CAR-iNKT cells was assessed by flow cytometry, image stream analysis and multiplex analysis in single-stimulation or repeated-stimulation assays. Moreover, the immunoregulatory properties of CD19-CAR-iNKT cells were analyzed in apoptosis assays and in mixed lymphocyte reactions. The effect of checkpoint inhibition through nivolumab was analyzed in these settings. RESULTS In this study, we could show that the cytotoxicity of CD19-CAR-iNKT cells was mediated either through engagement of their CAR or their invariant T-cell receptor, which may circumvent loss of response through antigen escape. However, encounter of CD19-CAR-iNKT cells with their target induced a phenotype of exhaustion. Consequently, checkpoint inhibition increased cytokine release, cytotoxicity and survival of CD19-CAR-iNKT cells. Additionally, they showed robust suppression of alloreactive immune responses. CONCLUSION In this work, we demonstrate that CAR-iNKT cells are a powerful cytotherapeutic option to prevent or treat relapse while potentially reducing the risk of GVHD after allogeneic HCT.
Collapse
Affiliation(s)
- Emmanuelle Moraes Ribeiro
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Kathy-Ann Secker
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Ana-Maria Nitulescu
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Rebekka Schairer
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Hildegard Keppeler
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Anton Wesle
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Hannes Schmid
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Anita Schmitt
- Department of Oncology, Hematology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Oncology, Hematology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniela Chmiest
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Silvia Podavini
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Boris Klimovich
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Michael Schmitt
- Department of Oncology, Hematology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fulya Korkmaz
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Claudia Lengerke
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Corina Schneidawind
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Dominik Schneidawind
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Yu Y, Zu L, Jiang J, Wu Y, Wang Y, Xu M, Liu Q. Structure-aware deep model for MHC-II peptide binding affinity prediction. BMC Genomics 2024; 25:127. [PMID: 38291350 PMCID: PMC10826266 DOI: 10.1186/s12864-023-09900-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/12/2023] [Indexed: 02/01/2024] Open
Abstract
The prediction of major histocompatibility complex (MHC)-peptide binding affinity is an important branch in immune bioinformatics, especially helpful in accelerating the design of disease vaccines and immunity therapy. Although deep learning-based solutions have yielded promising results on MHC-II molecules in recent years, these methods ignored structure knowledge from each peptide when employing the deep neural network models. Each peptide sequence has its specific combination order, so it is worth considering adding the structural information of the peptide sequence to the deep model training. In this work, we use positional encoding to represent the structural information of peptide sequences and validly combine the positional encoding with existing models by different strategies. Experiments on three datasets show that the introduction of position-coding information can further improve the performance built upon the existing model. The idea of introducing positional encoding to this field can provide important reference significance for the optimization of the deep network structure in the future.
Collapse
Affiliation(s)
- Ying Yu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Lipeng Zu
- Department of Computer Science, Florida State University, Tallahassee, 32306, USA
| | - Jiaye Jiang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yafang Wu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yinglin Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Midie Xu
- Department of Pathology, Fudan University, Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Institute of Pathology, Fudan University, Shanghai, 200032, China.
| | - Qing Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| |
Collapse
|
20
|
Maas-Bauer K, Köhler N, Stell AV, Zwick M, Acharya S, Rensing-Ehl A, König C, Kroll J, Baker J, Koßmann S, Pradier A, Wang S, Docquier M, Lewis DB, Negrin RS, Simonetta F. Single-cell transcriptomics reveal different maturation stages and sublineage commitment of human thymic invariant natural killer T cells. J Leukoc Biol 2024; 115:401-409. [PMID: 37742056 PMCID: PMC10799303 DOI: 10.1093/jleuko/qiad113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/08/2023] [Accepted: 08/29/2023] [Indexed: 09/25/2023] Open
Abstract
Invariant natural killer T cells are a rare, heterogeneous T-cell subset with cytotoxic and immunomodulatory properties. During thymic development, murine invariant natural killer T cells go through different maturation stages differentiating into distinct sublineages, namely, invariant natural killer T1, 2, and 17 cells. Recent reports indicate that invariant natural killer T2 cells display immature properties and give rise to other subsets, whereas invariant natural killer T1 cells seem to be terminally differentiated. Whether human invariant natural killer T cells follow a similar differentiation model is still unknown. To define the maturation stages and assess the sublineage commitment of human invariant natural killer T cells during thymic development, in this study, we performed single-cell RNA sequencing analysis on human Vα24+Vβ11+ invariant natural killer T cells isolated from thymocytes. We show that these invariant natural killer T cells displayed heterogeneity, and our unsupervised analysis identified 5 clusters representing different maturation stages, from an immature profile with high expression of genes important for invariant natural killer T cell development and proliferation to a mature, fully differentiated profile with high levels of cytotoxic effector molecules. Evaluation of expression of sublineage-defining gene sets revealed mainly cells with an invariant natural killer T2 signature in the most immature cluster, whereas the more differentiated ones displayed an invariant natural killer T1 signature. Combined analysis with a publicly available single-cell RNA sequencing data set of human invariant natural killer T cells from peripheral blood suggested that the 2 main subsets exist both in thymus and in the periphery, while a third more immature one was restricted to the thymus. Our data point to the existence of different maturation stages of human thymic invariant natural killer T cells and provide evidence for sublineage commitment of invariant natural killer T cells in the human thymus.
Collapse
Affiliation(s)
- Kristina Maas-Bauer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Center for Clinical Sciences Research Building, 269 W. Campus Drive, Stanford, CA 94305, United States
- Department of Hematology, Oncology, and Stem Cell Transplantation, Medical Center—University of Freiburg, Faculty of Medicine, Hugstetter Str. 55, Freiburg 79106, Germany
| | - Natalie Köhler
- Department of Hematology, Oncology, and Stem Cell Transplantation, Medical Center—University of Freiburg, Faculty of Medicine, Hugstetter Str. 55, Freiburg 79106, Germany
- CIBSS—Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestr. 18, Freiburg 79104, Germany
| | - Anna-Verena Stell
- Department of Hematology, Oncology, and Stem Cell Transplantation, Medical Center—University of Freiburg, Faculty of Medicine, Hugstetter Str. 55, Freiburg 79106, Germany
| | - Melissa Zwick
- Department of Hematology, Oncology, and Stem Cell Transplantation, Medical Center—University of Freiburg, Faculty of Medicine, Hugstetter Str. 55, Freiburg 79106, Germany
| | - Swati Acharya
- Sean N. Parker Center for Asthma and Allergy Research, Department of Medicine, Stanford University, 240 Pasteur Dr, Stanford, CA 94304, United States
| | - Anne Rensing-Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, Breisacher Str. 115, Freiburg 79106, Germany
| | - Christoph König
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, Breisacher Str. 115, Freiburg 79106, Germany
- Faculty of Biology, University of Freiburg, Schänzlestr. 1, Freiburg 79104, Germany
| | - Johannes Kroll
- Department of Cardiovascular Surgery, Heart Center Freiburg University, Hugstetter Straße 55, Freiburg 79106, Germany
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Center for Clinical Sciences Research Building, 269 W. Campus Drive, Stanford, CA 94305, United States
| | - Stefanie Koßmann
- Department of Hematology, Oncology, and Stem Cell Transplantation, Medical Center—University of Freiburg, Faculty of Medicine, Hugstetter Str. 55, Freiburg 79106, Germany
| | - Amandine Pradier
- Division of Hematology, Department of Oncology, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, Geneva 1205, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, Geneva 1211, Switzerland
| | - Sisi Wang
- Translational Research Center for Oncohematology, Department of Medicine, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, Geneva 1211, Switzerland
| | - Mylène Docquier
- iGE3 Genomics Platform, University of Geneva, Rue Michel-Servet 1, Geneva 1211, Switzerland
- Department of Genetics & Evolution, University of Geneva, Rue Michel-Servet 1, Geneva 1211, Switzerland
| | - David B Lewis
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, United States
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Center for Clinical Sciences Research Building, 269 W. Campus Drive, Stanford, CA 94305, United States
| | - Federico Simonetta
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Center for Clinical Sciences Research Building, 269 W. Campus Drive, Stanford, CA 94305, United States
- Division of Hematology, Department of Oncology, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, Geneva 1205, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, Geneva 1211, Switzerland
| |
Collapse
|
21
|
Zhao W, Li M, Song S, Zhi Y, Huan C, Lv G. The role of natural killer T cells in liver transplantation. Front Cell Dev Biol 2024; 11:1274361. [PMID: 38250325 PMCID: PMC10796773 DOI: 10.3389/fcell.2023.1274361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Natural killer T cells (NKTs) are innate-like lymphocytes that are abundant in the liver and participate in liver immunity. NKT cells express both NK cell and T cell markers, modulate innate and adaptive immune responses. Type I and Type II NKT cells are classified according to the TCR usage, while they recognize lipid antigen in a non-classical major histocompatibility (MHC) molecule CD1d-restricted manner. Once activated, NKT cells can quickly produce cytokines and chemokines to negatively or positively regulate the immune responses, depending on the different NKT subsets. In liver transplantation (LTx), the immune reactions in a series of processes determine the recipients' long-term survival, including ischemia-reperfusion injury, alloresponse, and post-transplant infection. This review provides insight into the research on NKT cells subpopulations in LTx immunity during different processes, and discusses the shortcomings of the current research on NKT cells. Additionally, the CD56-expressing T cells are recognized as a NK-like T cell population, they were also discussed during these processes.
Collapse
Affiliation(s)
- Wenchao Zhao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shifei Song
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yao Zhi
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chen Huan
- Center of Infectious Diseases and Pathogen Biology, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
22
|
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
|
23
|
Textor J, Buytenhuijs F, Rogers D, Gauthier ÈM, Sultan S, Wortel IMN, Kalies K, Fähnrich A, Pagel R, Melichar HJ, Westermann J, Mandl JN. Machine learning analysis of the T cell receptor repertoire identifies sequence features of self-reactivity. Cell Syst 2023; 14:1059-1073.e5. [PMID: 38061355 DOI: 10.1016/j.cels.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/01/2023] [Accepted: 11/09/2023] [Indexed: 12/23/2023]
Abstract
The T cell receptor (TCR) determines specificity and affinity for both foreign and self-peptides presented by the major histocompatibility complex (MHC). Although the strength of TCR interactions with self-pMHC impacts T cell function, it has been challenging to identify TCR sequence features that predict T cell fate. To discern patterns distinguishing TCRs from naive CD4+ T cells with low versus high self-reactivity, we used data from 42 mice to train a machine learning (ML) algorithm that identifies population-level differences between TCRβ sequence sets. This approach revealed that weakly self-reactive T cell populations were enriched for longer CDR3β regions and acidic amino acids. We tested our ML predictions of self-reactivity using retrogenic mice with fixed TCRβ sequences. Extrapolating our analyses to independent datasets, we predicted high self-reactivity for regulatory T cells and slightly reduced self-reactivity for T cells responding to chronic infections. Our analyses suggest a potential trade-off between TCR repertoire diversity and self-reactivity. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Johannes Textor
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, the Netherlands; Medical BioSciences, Radboudumc, Nijmegen 6525 GA, the Netherlands.
| | - Franka Buytenhuijs
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, the Netherlands
| | - Dakota Rogers
- Department of Physiology, McGill University, Montreal, QC H3G 0B1, Canada; McGill Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada
| | - Ève Mallet Gauthier
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Shabaz Sultan
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, the Netherlands; Medical BioSciences, Radboudumc, Nijmegen 6525 GA, the Netherlands
| | - Inge M N Wortel
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, the Netherlands; Medical BioSciences, Radboudumc, Nijmegen 6525 GA, the Netherlands
| | - Kathrin Kalies
- Institut für Anatomie, Universität zu Lübeck, 23562 Lübeck, Germany
| | - Anke Fähnrich
- Institut für Anatomie, Universität zu Lübeck, 23562 Lübeck, Germany
| | - René Pagel
- Institut für Anatomie, Universität zu Lübeck, 23562 Lübeck, Germany
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada; Department of Medicine, Université de Montréal, Montréal, QC H1T 2M4, Canada; Department of Microbiology & Immunology, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | | | - Judith N Mandl
- Department of Physiology, McGill University, Montreal, QC H3G 0B1, Canada; Department of Microbiology & Immunology, McGill University, Montreal, QC H3A 1A3, Canada; McGill Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada.
| |
Collapse
|
24
|
Poddighe D, Maulenkul T, Zhubanova G, Akhmaldtinova L, Dossybayeva K. Natural Killer T (NKT) Cells in Autoimmune Hepatitis: Current Evidence from Basic and Clinical Research. Cells 2023; 12:2854. [PMID: 38132174 PMCID: PMC10742140 DOI: 10.3390/cells12242854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Natural killer T (NKT) cells are unconventional T cells that are activated by glycolipid antigens. They can produce a variety of inflammatory and regulatory cytokines and, therefore, modulate multiple aspects of the immune response in different pathological settings, including autoimmunity. NKT cells have also been implicated in the immunopathogenesis of autoimmune hepatitis, and in this review we summarize and analyze the main studies investigating the involvement and/or homeostasis of NKT cells in this disease. In detail, the evidence from both basic and clinical research has been specifically analyzed. Even though the experimental murine models supported a relevant role of NKT cells in immune-mediated hepatic injury, very few studies specifically investigated NKT cell homeostasis in patients with autoimmune hepatitis; however, these initial studies reported some alterations of NKT cells in these patients, which may also correlate with the disease activity to some extent. Further clinical studies are needed to investigate the potential role and use of NKT cell analysis as a disease marker of clinical relevance, and to better understand the precise cellular and molecular mechanisms by which NKT cells contribute to the pathogenesis of autoimmune hepatitis.
Collapse
Affiliation(s)
- Dimitri Poddighe
- School of Medicine, Nazarbayev University, Kerei-Zhanibek Str. 5/1, Astana 010000, Kazakhstan
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana 010000, Kazakhstan
| | - Tilektes Maulenkul
- School of Medicine, Nazarbayev University, Kerei-Zhanibek Str. 5/1, Astana 010000, Kazakhstan
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana 010000, Kazakhstan
| | - Gulsamal Zhubanova
- School of Medicine, Nazarbayev University, Kerei-Zhanibek Str. 5/1, Astana 010000, Kazakhstan
| | - Lyudmila Akhmaldtinova
- School of Medicine, Nazarbayev University, Kerei-Zhanibek Str. 5/1, Astana 010000, Kazakhstan
| | - Kuanysh Dossybayeva
- School of Medicine, Nazarbayev University, Kerei-Zhanibek Str. 5/1, Astana 010000, Kazakhstan
| |
Collapse
|
25
|
Tyrrell DJ, Wragg KM, Chen J, Wang H, Song J, Blin MG, Bolding C, Vardaman D, Giles K, Tidwell H, Ali MA, Janappareddi A, Wood SC, Goldstein DR. Clonally expanded memory CD8 + T cells accumulate in atherosclerotic plaques and are pro-atherogenic in aged mice. NATURE AGING 2023; 3:1576-1590. [PMID: 37996758 PMCID: PMC11924142 DOI: 10.1038/s43587-023-00515-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/27/2023] [Indexed: 11/25/2023]
Abstract
Aging is a strong risk factor for atherosclerosis and induces accumulation of memory CD8+ T cells in mice and humans. Biological changes that occur with aging lead to enhanced atherosclerosis, yet the role of aging on CD8+ T cells during atherogenesis is unclear. In this study, using femle mice, we found that depletion of CD8+ T cells attenuated atherogenesis in aged, but not young, animals. Furthermore, adoptive transfer of splenic CD8+ T cells from aged wild-type, but not young wild-type, donor mice significantly enhanced atherosclerosis in recipient mice lacking CD8+ T cells. We also characterized T cells in healthy and atherosclerotic young and aged mice by single-cell RNA sequencing. We found specific subsets of age-associated CD8+ T cells, including a Granzyme K+ effector memory subset, that accumulated and was clonally expanded within atherosclerotic plaques. These had transcriptomic signatures of T cell activation, migration, cytotoxicity and exhaustion. Overall, our study identified memory CD8+ T cells as therapeutic targets for atherosclerosis in aging.
Collapse
Affiliation(s)
- Daniel J Tyrrell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Kathleen M Wragg
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Judy Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Hui Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jianrui Song
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Muriel G Blin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chase Bolding
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donald Vardaman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kara Giles
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Harrison Tidwell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Md Akkas Ali
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Sherri C Wood
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Daniel R Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Amable L, Ferreira Martins LA, Pierre R, Do Cruseiro M, Chabab G, Sergé A, Kergaravat C, Delord M, Viret C, Jaubert J, Liu C, Karray S, Marie JC, Irla M, Georgiev H, Clave E, Toubert A, Lucas B, Klibi J, Benlagha K. Intrinsic factors and CD1d1 but not CD1d2 expression levels control invariant natural killer T cell subset differentiation. Nat Commun 2023; 14:7922. [PMID: 38040679 PMCID: PMC10692182 DOI: 10.1038/s41467-023-43424-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 11/08/2023] [Indexed: 12/03/2023] Open
Abstract
Invariant natural killer T (NKT) cell subsets are defined based on their cytokine-production profiles and transcription factors. Their distribution is different in C57BL/6 (B6) and BALB/c mice, with a bias for NKT1 and NKT2/NKT17 subsets, respectively. Here, we show that the non-classical class I-like major histocompatibility complex CD1 molecules CD1d2, expressed in BALB/c and not in B6 mice, could not account for this difference. We find however that NKT cell subset distribution is intrinsic to bone marrow derived NKT cells, regardless of syngeneic CD1d-ligand recognition, and that multiple intrinsic factors are likely involved. Finally, we find that CD1d expression levels in combination with T cell antigen receptor signal strength could also influence NKT cell distribution and function. Overall, this study indicates that CD1d-mediated TCR signals and other intrinsic signals integrate to influence strain-specific NKT cell differentiation programs and subset distributions.
Collapse
Affiliation(s)
- Ludivine Amable
- Université Paris Cité, Institut de Recherche Saint-Louis (IRSL), EMiLy, Paris, France
| | | | - Remi Pierre
- Plateforme de recombinaison homologue et de cryoconservation (PRHTEC), Institut Cochin, Université Paris Descartes, Paris, France
| | - Marcio Do Cruseiro
- Plateforme de recombinaison homologue et de cryoconservation (PRHTEC), Institut Cochin, Université Paris Descartes, Paris, France
| | - Ghita Chabab
- Tumor Escape Resistance and Immunity department, Cancer Research Center of Lyon INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Lyon, France
| | - Arnauld Sergé
- Laboratoire Adhésion Inflammation (LAI), CNRS, INSERM, Aix-Marseille Université, Marseille, France
| | - Camille Kergaravat
- Université Paris Cité, Institut de Recherche Saint-Louis (IRSL), EMiLy, Paris, France
| | | | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Jean Jaubert
- Mouse Genetics Unit, Institut Pasteur, Paris, France
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science Technology, Wuhan, China
| | - Saoussen Karray
- Université Paris Cité, INSERM U976, Institut de Recherche Saint-Louis (IRSL), Hôpital Saint-Louis, Paris, France
| | - Julien C Marie
- Tumor Escape Resistance and Immunity department, Cancer Research Center of Lyon INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Lyon, France
| | - Magali Irla
- Centre d'Immunologie de Marseille-Luminy (CIML), CNRS, INSERM, Aix-Marseille Université, Marseille, France
| | - Hristo Georgiev
- Institute of immunology, Hannover Medical School, Hannover, Germany
| | - Emmanuel Clave
- Université Paris Cité, Institut de Recherche Saint-Louis (IRSL), EMiLy, Paris, France
| | - Antoine Toubert
- Université Paris Cité, Institut de Recherche Saint-Louis (IRSL), EMiLy, Paris, France
| | - Bruno Lucas
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Université Paris Descartes, Paris, France
| | - Jihene Klibi
- Université Paris Cité, Institut de Recherche Saint-Louis (IRSL), EMiLy, Paris, France
| | - Kamel Benlagha
- Université Paris Cité, Institut de Recherche Saint-Louis (IRSL), EMiLy, Paris, France.
| |
Collapse
|
27
|
Chengalroyen MD. Current Perspectives and Challenges of MAIT Cell-Directed Therapy for Tuberculosis Infection. Pathogens 2023; 12:1343. [PMID: 38003807 PMCID: PMC10675005 DOI: 10.3390/pathogens12111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/27/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are a distinct population of non-conventional T cells that have been preserved through evolution and possess properties of both innate and adaptive immune cells. They are activated through the recognition of antigens presented by non-polymorphic MR1 proteins or, alternately, can be stimulated by specific cytokines. These cells are multifaceted and exert robust antimicrobial activity against bacterial and viral infections, direct the immune response through the modulation of other immune cells, and exhibit a specialized tissue homeostasis and repair function. These distinct characteristics have instigated interest in MAIT cell biology for immunotherapy and vaccine development. This review describes the current understanding of MAIT cell activation, their role in infections and diseases with an emphasis on tuberculosis (TB) infection, and perspectives on the future use of MAIT cells in immune-mediated therapy.
Collapse
Affiliation(s)
- Melissa D Chengalroyen
- Molecular Mycobacteriology Research Unit, Institute of Infectious Disease and Molecular Medicine, Department of Pathology, University of Cape Town, Cape Town 7700, South Africa
| |
Collapse
|
28
|
Zappa E, Vitali A, Anders K, Molenaar JJ, Wienke J, Künkele A. Adoptive cell therapy in paediatric extracranial solid tumours: current approaches and future challenges. Eur J Cancer 2023; 194:113347. [PMID: 37832507 PMCID: PMC10695178 DOI: 10.1016/j.ejca.2023.113347] [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: 07/17/2023] [Revised: 09/03/2023] [Accepted: 09/09/2023] [Indexed: 10/15/2023]
Abstract
Immunotherapy has ignited hope to cure paediatric solid tumours that resist traditional therapies. Among the most promising methods is adoptive cell therapy (ACT). Particularly, ACT using T cells equipped with chimeric antigen receptors (CARs) has moved into the spotlight in clinical studies. However, the efficacy of ACT is challenged by ACT-intrinsic factors, like lack of activation or T cell exhaustion, as well as immune evasion strategies of paediatric solid tumours, such as their highly immunosuppressive microenvironment. Novel strategies, including ACT using innate-like lymphocytes, innovative cell engineering techniques, and ACT combination therapies, are being developed and will be crucial to overcome these challenges. Here, we discuss the main classes of ACT for the treatment of paediatric extracranial solid tumours, reflect on the available preclinical and clinical evidence supporting promising strategies, and address the challenges that ACT is still facing. Ultimately, we highlight state-of-the-art developments and opportunities for new therapeutic options, which hold great potential for improving outcomes in this challenging patient population.
Collapse
Affiliation(s)
- Elisa Zappa
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Alice Vitali
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany.
| | - Kathleen Anders
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Judith Wienke
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Annette Künkele
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
29
|
Lee MS, Webb TJ. Novel lipid antigens for NKT cells in cancer. Front Immunol 2023; 14:1173375. [PMID: 37908366 PMCID: PMC10613688 DOI: 10.3389/fimmu.2023.1173375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Cancer immunotherapy aims to unleash the power of the immune system against tumors without the side effects of traditional chemotherapy. Immunotherapeutic methods vary widely, but all follow the same basic principle: overcome the barriers utilized by cancers to avoid immune destruction. These approaches often revolve around classical T cells, such as with CAR T cells and neoantigen vaccines; however, the utility of the innate-like iNKT cell in cancer immunotherapy has gained significant recognition. iNKT cells parallel classic T cell recognition of peptide antigens presented on MHC through their recognition of lipid antigens presented on the MHC I-like molecule CD1d. Altered metabolism and a lipogenic phenotype are essential properties of tumor cells, representing a unique feature that may be exploited by iNKT cells. In this review, we will cover properties of iNKT cells, CD1d, and lipid antigen presentation. Next, we will discuss the cancer lipidome and how it may be exploited by iNKT cells through a window of opportunity. Finally, we will review, in detail, novel lipid antigens for iNKT cells in cancer.
Collapse
Affiliation(s)
- Michael S. Lee
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tonya J. Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
30
|
Kurioka A, Klenerman P. Aging unconventionally: γδ T cells, iNKT cells, and MAIT cells in aging. Semin Immunol 2023; 69:101816. [PMID: 37536148 PMCID: PMC10804939 DOI: 10.1016/j.smim.2023.101816] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023]
Abstract
Unconventional T cells include γδ T cells, invariant Natural Killer T cells (iNKT) cells and Mucosal Associated Invariant T (MAIT) cells, which are distinguished from conventional T cells by their recognition of non-peptide ligands presented by non-polymorphic antigen presenting molecules and rapid effector functions that are pre-programmed during their development. Here we review current knowledge of the effect of age on unconventional T cells, from early life to old age, in both mice and humans. We then discuss the role of unconventional T cells in age-associated diseases and infections, highlighting the similarities between members of the unconventional T cell family in the context of aging.
Collapse
Affiliation(s)
- Ayako Kurioka
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Cameron G, Nguyen T, Ciula M, Williams SJ, Godfrey DI. Glycolipids from the gut symbiont Bacteroides fragilis are agonists for natural killer T cells and induce their regulatory differentiation. Chem Sci 2023; 14:7887-7896. [PMID: 37502334 PMCID: PMC10370605 DOI: 10.1039/d3sc02124f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 07/29/2023] Open
Abstract
Natural Killer T (NKT) cells are a lipid-antigen reactive T cell subset that is restricted to the antigen presenting molecule CD1d. They possess diverse functional properties that contribute to inflammatory and regulatory immune responses. The most studied lipid antigen target for these T cells is α-galactosylceramide (αGC). The commensal organism Bacteroides fragilis (B. fragilis) produces several forms of αGC, but conflicting information exists about the influence of these lipids on NKT cells. Herein, we report the total synthesis of a major form of αGC from B. fragilis (Bf αGC), and several analogues thereof. We confirm the T cell receptor (TCR)-mediated recognition of these glycolipids by mouse and human NKT cells. Despite the natural structure of Bf αGC containing lipid branching that limits potency, we demonstrate that Bf αGC drives mouse NKT cells to proliferate and differentiate into producers of the immunoregulatory cytokine, interleukin-10 (IL-10). These Bf αGC-experienced NKT cells display regulatory function by inhibiting the expansion of naïve NKT cells upon subsequent exposure to this antigen. Moreover, this regulatory activity impacts more than just NKT cells, as demonstrated by the NKT cell-mediated inhibition of antigen-stimulated mucosal-associated invariant T (MAIT) cells (a T cell subset restricted to a different antigen presenting molecule, MR1). These findings reveal that B. fragilis-derived NKT cell agonists may have broad immunoregulatory activity, providing insight into the mechanisms influencing immune tolerance to commensal bacteria and highlighting a potential means to manipulate NKT cell function for therapeutic benefit.
Collapse
Affiliation(s)
- Garth Cameron
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| | - Tram Nguyen
- School of Chemistry, University of Melbourne Parkville VIC 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne Parkville VIC 3010 Australia
| | - Marcin Ciula
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| | - Spencer J Williams
- School of Chemistry, University of Melbourne Parkville VIC 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne Parkville VIC 3010 Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| |
Collapse
|
32
|
Lagattuta KA, Nathan A, Rumker L, Birnbaum ME, Raychaudhuri S. The T cell receptor sequence influences the likelihood of T cell memory formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.20.549939. [PMID: 37502994 PMCID: PMC10370203 DOI: 10.1101/2023.07.20.549939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
T cell differentiation depends on activation through the T cell receptor (TCR), whose amino acid sequence varies cell to cell. Particular TCR amino acid sequences nearly guarantee Mucosal-Associated Invariant T (MAIT) and Natural Killer T (NKT) cell fates. To comprehensively define how TCR amino acids affects all T cell fates, we analyze the paired αβTCR sequence and transcriptome of 819,772 single cells. We find that hydrophobic CDR3 residues promote regulatory T cell transcriptional states in both the CD8 and CD4 lineages. Most strikingly, we find a set of TCR sequence features, concentrated in CDR2α, that promotes positive selection in the thymus as well as transition from naïve to memory in the periphery. Even among T cells that recognize the same antigen, these TCR sequence features help to explain which T cells form immunological memory, which is essential for effective pathogen response.
Collapse
Affiliation(s)
- Kaitlyn A. Lagattuta
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Laurie Rumker
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael E. Birnbaum
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Wang NI, Ninkov M, Haeryfar SMM. Classic costimulatory interactions in MAIT cell responses: from gene expression to immune regulation. Clin Exp Immunol 2023; 213:50-66. [PMID: 37279566 PMCID: PMC10324557 DOI: 10.1093/cei/uxad061] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/17/2023] [Accepted: 06/01/2023] [Indexed: 06/08/2023] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are evolutionarily conserved, innate-like T lymphocytes with enormous immunomodulatory potentials. Due to their strategic localization, their invariant T cell receptor (iTCR) specificity for major histocompatibility complex-related protein 1 (MR1) ligands of commensal and pathogenic bacterial origin, and their sensitivity to infection-elicited cytokines, MAIT cells are best known for their antimicrobial characteristics. However, they are thought to also play important parts in the contexts of cancer, autoimmunity, vaccine-induced immunity, and tissue repair. While cognate MR1 ligands and cytokine cues govern MAIT cell maturation, polarization, and peripheral activation, other signal transduction pathways, including those mediated by costimulatory interactions, regulate MAIT cell responses. Activated MAIT cells exhibit cytolytic activities and secrete potent inflammatory cytokines of their own, thus transregulating the biological behaviors of several other cell types, including dendritic cells, macrophages, natural killer cells, conventional T cells, and B cells, with significant implications in health and disease. Therefore, an in-depth understanding of how costimulatory pathways control MAIT cell responses may introduce new targets for optimized MR1/MAIT cell-based interventions. Herein, we compare and contrast MAIT cells and mainstream T cells for their expression of classic costimulatory molecules belonging to the immunoglobulin superfamily and the tumor necrosis factor (TNF)/TNF receptor superfamily, based not only on the available literature but also on our transcriptomic analyses. We discuss how these molecules participate in MAIT cells' development and activities. Finally, we introduce several pressing questions vis-à-vis MAIT cell costimulation and offer new directions for future research in this area.
Collapse
Affiliation(s)
- Nicole I Wang
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Marina Ninkov
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Ontario, Canada
- Division of General Surgery, Department of Surgery, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
34
|
Rashu R, Ninkov M, Wardell CM, Benoit JM, Wang NI, Meilleur CE, D'Agostino MR, Zhang A, Feng E, Saeedian N, Bell GI, Vahedi F, Hess DA, Barr SD, Troyer RM, Kang CY, Ashkar AA, Miller MS, Haeryfar SMM. Targeting the MR1-MAIT cell axis improves vaccine efficacy and affords protection against viral pathogens. PLoS Pathog 2023; 19:e1011485. [PMID: 37384813 DOI: 10.1371/journal.ppat.1011485] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are MR1-restricted, innate-like T lymphocytes with tremendous antibacterial and immunomodulatory functions. Additionally, MAIT cells sense and respond to viral infections in an MR1-independent fashion. However, whether they can be directly targeted in immunization strategies against viral pathogens is unclear. We addressed this question in multiple wild-type and genetically altered but clinically relevant mouse strains using several vaccine platforms against influenza viruses, poxviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We demonstrate that 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU), a riboflavin-based MR1 ligand of bacterial origin, can synergize with viral vaccines to expand MAIT cells in multiple tissues, reprogram them towards a pro-inflammatory MAIT1 phenotype, license them to bolster virus-specific CD8+ T cell responses, and potentiate heterosubtypic anti-influenza protection. Repeated 5-OP-RU administration did not render MAIT cells anergic, thus allowing for its inclusion in prime-boost immunization protocols. Mechanistically, tissue MAIT cell accumulation was due to their robust proliferation, as opposed to altered migratory behavior, and required viral vaccine replication competency and Toll-like receptor 3 and type I interferon receptor signaling. The observed phenomenon was reproducible in female and male mice, and in both young and old animals. It could also be recapitulated in a human cell culture system in which peripheral blood mononuclear cells were exposed to replicating virions and 5-OP-RU. In conclusion, although viruses and virus-based vaccines are devoid of the riboflavin biosynthesis machinery that supplies MR1 ligands, targeting MR1 enhances the efficacy of vaccine-elicited antiviral immunity. We propose 5-OP-RU as a non-classic but potent and versatile vaccine adjuvant against respiratory viruses.
Collapse
Affiliation(s)
- Rasheduzzaman Rashu
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Marina Ninkov
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Christine M Wardell
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Jenna M Benoit
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Nicole I Wang
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Courtney E Meilleur
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Michael R D'Agostino
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Ali Zhang
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Emily Feng
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Nasrin Saeedian
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Gillian I Bell
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Fatemeh Vahedi
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - David A Hess
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Stephen D Barr
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Ryan M Troyer
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Chil-Yong Kang
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Ali A Ashkar
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Ontario, Canada
- Division of General Surgery, Department of Surgery, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
35
|
Ligthart NAM, de Geus MAR, van de Plassche MAT, Torres García D, Isendoorn MME, Reinalda L, Ofman D, van Leeuwen T, van Kasteren SI. A Lysosome-Targeted Tetrazine for Organelle-Specific Click-to-Release Chemistry in Antigen Presenting Cells. J Am Chem Soc 2023. [PMID: 37269296 DOI: 10.1021/jacs.3c02139] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Bioorthogonal deprotections are readily used to control biological function in a cell-specific manner. To further improve the spatial resolution of these reactions, we here present a lysosome-targeted tetrazine for an organelle-specific deprotection reaction. We show that trans-cyclooctene deprotection with this reagent can be used to control the biological activity of ligands for invariant natural killer T cells in the lysosome to shed light on the processing pathway in antigen presenting cells. We then use the lysosome-targeted tetrazine to show that long peptide antigens used for CD8+ T cell activation do not pass through this organelle, suggesting a role for the earlier endosomal compartments for their processing.
Collapse
Affiliation(s)
- Nina A M Ligthart
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Mark A R de Geus
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Merel A T van de Plassche
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Diana Torres García
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Marjolein M E Isendoorn
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Luuk Reinalda
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Daniëlle Ofman
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Tyrza van Leeuwen
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Sander I van Kasteren
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
36
|
Liman N, Park JH. Markers and makers of NKT17 cells. Exp Mol Med 2023; 55:1090-1098. [PMID: 37258582 PMCID: PMC10317953 DOI: 10.1038/s12276-023-01015-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 06/02/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are thymus-generated innate-like αβ T cells that undergo terminal differentiation in the thymus. Such a developmental pathway differs from that of conventional αβ T cells, which are generated in the thymus but complete their functional maturation in peripheral tissues. Multiple subsets of iNKT cells have been described, among which IL-17-producing iNKT cells are commonly referred to as NKT17 cells. IL-17 is considered a proinflammatory cytokine that can play both protective and pathogenic roles and has been implicated as a key regulatory factor in many disease settings. Akin to other iNKT subsets, NKT17 cells acquire their effector function during thymic development. However, the cellular mechanisms that drive NKT17 subset specification, and how iNKT cells in general acquire their effector function prior to antigen encounter, remain largely unknown. Considering that all iNKT cells express the canonical Vα14-Jα18 TCRα chain and all iNKT subsets display the same ligand specificity, i.e., glycolipid antigens in the context of the nonclassical MHC-I molecule CD1d, the conundrum is explaining how thymic NKT17 cell specification is determined. Mapping of the molecular circuitry of NKT17 cell differentiation, combined with the discovery of markers that identify NKT17 cells, has provided new insights into the developmental pathway of NKT17 cells. The current review aims to highlight recent advances in our understanding of thymic NKT17 cell development and to place these findings in the larger context of iNKT subset specification and differentiation.
Collapse
Affiliation(s)
- Nurcin Liman
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
37
|
Ikazaki T, Ishikawa E, Tamashima H, Akiyama H, Kimuro Y, Yoritate M, Matoba H, Imamura A, Ishida H, Yamasaki S, Hirai G. Ligand-Controlled Stereoselective Synthesis and Biological Activity of 2-Exomethylene Pseudo-glycoconjugates: Discovery of Mincle-Selective Ligands. Angew Chem Int Ed Engl 2023; 62:e202302569. [PMID: 37005509 DOI: 10.1002/anie.202302569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023]
Abstract
Glycoconjugate analogues in which the sp3 -hybridized C2 position of the carbohydrate structure (normally bearing a hydroxy group) is converted into a compact sp2 -hybridized exomethylene group are expected to have unique biological activities. We established ligand-controlled Tsuji-Trost-type glycosylation methodology to directly prepare a variety of these 2-exomethylene pseudo-glycoconjugates, including glucosylceramide analogues, in an α- or β-selective manner. Glucocerebrosidase GBA1 cleaves these synthetic pseudo-β-glucosylceramides similarly to native glucosylceramides. The pseudo-glucosylceramides exhibit selective ligand activity towards macrophage-inducible C-type lectin (Mincle), but unlike native glucosylceramides, are inactive towards CD1d.
Collapse
Affiliation(s)
- Takahiro Ikazaki
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroto Tamashima
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hisako Akiyama
- Juntendo Advanced Research Institute for Health Science, Juntendo University, Tokyo, 113-8421, Japan
- RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Yusuke Kimuro
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Makoto Yoritate
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroaki Matoba
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akihiro Imamura
- Department of Applied Bioorganic Chemistry and Institute for Glyco-core Research (iGCORE), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Hideharu Ishida
- Department of Applied Bioorganic Chemistry and Institute for Glyco-core Research (iGCORE), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Go Hirai
- Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
38
|
Ikazaki T, Ishikawa E, Tamashima H, Akiyama H, Kimuro Y, Yoritate M, Matoba H, Imamura A, Ishida H, Yamasaki S, Hirai G. Ligand‐Controlled Stereoselective Synthesis and Biological Activity of 2‐Exomethylene Pseudo‐glycoconjugates: Discovery of Mincle‐Selective Ligands. Angew Chem Int Ed Engl 2023; 135. [DOI: 10.1002/ange.202302569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Indexed: 04/05/2023]
Abstract
AbstractGlycoconjugate analogues in which the sp3‐hybridized C2 position of the carbohydrate structure (normally bearing a hydroxy group) is converted into a compact sp2‐hybridized exomethylene group are expected to have unique biological activities. We established ligand‐controlled Tsuji–Trost‐type glycosylation methodology to directly prepare a variety of these 2‐exomethylene pseudo‐glycoconjugates, including glucosylceramide analogues, in an α‐ or β‐selective manner. Glucocerebrosidase GBA1 cleaves these synthetic pseudo‐β‐glucosylceramides similarly to native glucosylceramides. The pseudo‐glucosylceramides exhibit selective ligand activity towards macrophage‐inducible C‐type lectin (Mincle), but unlike native glucosylceramides, are inactive towards CD1d.
Collapse
Affiliation(s)
- Takahiro Ikazaki
- Graduate School of Pharmaceutical Sciences Kyushu University Maidashi Higashi-ku Fukuoka 812-8582 Japan
| | - Eri Ishikawa
- Department of Molecular Immunology Research Institute for Microbial Diseases Osaka University Yamadaoka Suita Osaka 565-0871 Japan
- Laboratory of Molecular Immunology Immunology Frontier Research Center Osaka University Yamadaoka Suita Osaka 565-0871 Japan
| | - Hiroto Tamashima
- Graduate School of Pharmaceutical Sciences Kyushu University Maidashi Higashi-ku Fukuoka 812-8582 Japan
| | - Hisako Akiyama
- Juntendo Advanced Research Institute for Health Science Juntendo University Tokyo 113-8421 Japan
- RIKEN Center for Brain Science Wako Saitama 351-0198 Japan
| | - Yusuke Kimuro
- Graduate School of Pharmaceutical Sciences Kyushu University Maidashi Higashi-ku Fukuoka 812-8582 Japan
| | - Makoto Yoritate
- Graduate School of Pharmaceutical Sciences Kyushu University Maidashi Higashi-ku Fukuoka 812-8582 Japan
| | - Hiroaki Matoba
- Graduate School of Pharmaceutical Sciences Kyushu University Maidashi Higashi-ku Fukuoka 812-8582 Japan
| | - Akihiro Imamura
- Department of Applied Bioorganic Chemistry and Institute for Glyco-core Research (iGCORE) Gifu University 1-1 Yanagido Gifu 501-1193 Japan
| | - Hideharu Ishida
- Department of Applied Bioorganic Chemistry and Institute for Glyco-core Research (iGCORE) Gifu University 1-1 Yanagido Gifu 501-1193 Japan
| | - Sho Yamasaki
- Department of Molecular Immunology Research Institute for Microbial Diseases Osaka University Yamadaoka Suita Osaka 565-0871 Japan
- Laboratory of Molecular Immunology Immunology Frontier Research Center Osaka University Yamadaoka Suita Osaka 565-0871 Japan
| | - Go Hirai
- Graduate School of Pharmaceutical Sciences Kyushu University Maidashi Higashi-ku Fukuoka 812-8582 Japan
| |
Collapse
|
39
|
Aoki T, Motohashi S, Koseki H. Regeneration of invariant natural killer T (iNKT) cells: application of iPSC technology for iNKT cell-targeted tumor immunotherapy. Inflamm Regen 2023; 43:27. [PMID: 37170375 PMCID: PMC10176773 DOI: 10.1186/s41232-023-00275-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/29/2023] [Indexed: 05/13/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a subset of innate-like T cells restricted by a major histocompatibility complex (MHC) class I-like molecule, CD1d. iNKT cells express an invariant T cell receptor (TCR) encoded by Vα14 Jα18 in mice and Vα24 Jα18 in humans and are activated by recognizing glycolipid antigens, such as α-galactosylceramide (αGalCer), presented by CD1d. iNKT cells exhibit anti-tumor activity via their NK-like cytotoxicity and adjuvant activity. Although iNKT cell-targeted immunotherapy is a conceptually promising approach, we still found a technical hurdle for its clinical implementation which is mainly due to the low frequency of iNKT cells, particularly in humans. To compensate for this, we proposed to generate adequate numbers of clinically competent NKT cells from induced pluripotent stem cells (iPSCs) for cancer immunotherapy. Toward this goal, we first obtained the proof of concept (POC) for this approach in mice. We developed a technology to differentiate iPSCs into iNKT cells (iPSC-iNKT cells) and found iPSC-iNKT cells efficiently rejected a syngeneic experimental thymoma by inducing antigen-specific CD8 T cells. After achieving the POC in mice, we developed human iPSC-iNKT cells, which had a high correlation in their gene expression profiles with parental iNKT cells. Human iPSC-iNKT cells also exhibited anti-tumor activity and adjuvant activity for human NK cells in vivo. Based on this supporting evidence for the anti-tumor activity of human iPSC-iNKT cells, we began to generate good manufacturing practice (GMP)-grade iPSC-iNKT cells. As of now, the first-in-human clinical trial of iPSC-iNKT cell therapy is ongoing as a single-agent, dose-escalation study for patients with advanced head and neck cancer. Demonstration of the safety of iPSC-iNKT cell therapy may allow us to improve the strategy by further reinforcing the therapeutic activity of iPSC-iNKT, cells either by gene-editing or combinatorial use with other immune cell products such as dendritic cells. Sixteen years after the establishment of the iPSC technology, we are reaching the first checkpoint to evaluate the clinical efficacy of iPSC-derived immune cells.
Collapse
Affiliation(s)
- Takahiro Aoki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Department of Medical Immunology, 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, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8670, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
40
|
Tsumura R, Haruta M, Kuwano M, Yasunaga M. Expansion of mixed immune cells using CD3/CD161 co-stimulation for the treatment of cancer. Sci Rep 2023; 13:6803. [PMID: 37100864 PMCID: PMC10133288 DOI: 10.1038/s41598-023-33987-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 04/21/2023] [Indexed: 04/28/2023] Open
Abstract
Adoptive cell transfer (ACT) is a type of personalized immunotherapy in which expanded immune cells are administered to patients with cancer. However, single-cell populations, such as killer T cells, dendritic cells, natural killer (NK) cells, and NKT (NKT) cells, have been generally used, and their effectiveness remains limited. Here, we established a novel culture method via CD3/CD161 co-stimulation and successfully expanded CD3+/CD4+ helper T cells, CD3+/CD8+ cytotoxic T cells (CTLs), CD3-/CD56+ NK cells, CD3+/CD1d+ NKT cells, CD3+/CD56+ NKT cells, CD3+/TCRγδ+ T cells, and CD3-/CD11c+/HLA-DR+ dendritic cells in peripheral blood mononuclear cells from healthy donors; their respective numbers were 155.5, 1132.5, 5.7, 117.0, 659.2, 325.6, and 6.8 times higher than those before expansion. These mixed immune cells showed strong cytotoxicity against cancer cell lines Capan-1 and SW480. Moreover, both CD3+/CD8+ CTLs and CD3+/CD56+ NKT cells killed tumor cells in cell contact-dependent and -independent manners via granzyme B and interferon-γ/TNF-α, respectively. Furthermore, the cytotoxicity of the mixed cells was significantly superior to that of CTLs or NKTs alone. A bet-hedging CTL-NKT circuitry is one potential mechanism underlying this cooperative cytotoxicity. Collectively, CD3/CD161 co-stimulation may be a promising culture method to expand multiple, distinct immune cell populations for the treatment of cancer.
Collapse
Affiliation(s)
- Ryo Tsumura
- Division of Developmental Therapeutics, EPOC, National Cancer Center, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Miwa Haruta
- Dojin institute of cancer immunology, Co., Ltd. Kumamoto, Kumamoto, 862-0967, Japan
| | - Masataka Kuwano
- Dojin institute of cancer immunology, Co., Ltd. Kumamoto, Kumamoto, 862-0967, Japan
| | - Masahiro Yasunaga
- Division of Developmental Therapeutics, EPOC, National Cancer Center, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
41
|
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
|
42
|
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
|
43
|
Lee SK, Choi JY, Jung ES, Kwon JH, Jang JW, Bae SH, Yoon SK. An Immunological Perspective on the Mechanism of Drug Induced Liver Injury: Focused on Drugs for Treatment of Hepatocellular Carcinoma and Liver Transplantation. Int J Mol Sci 2023; 24:5002. [PMID: 36902432 PMCID: PMC10003078 DOI: 10.3390/ijms24055002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
The liver is frequently exposed to potentially toxic materials, and it is the primary site of clearance of foreign agents, along with many innate and adaptive immune cells. Subsequently, drug induced liver injury (DILI), which is caused by medications, herbs, and dietary supplements, often occurs and has become an important issue in liver diseases. Reactive metabolites or drug-protein complexes induce DILI via the activation of various innate and adaptive immune cells. There has been a revolutionary development of treatment drugs for hepatocellular carcinoma (HCC) and liver transplantation (LT), including immune checkpoint inhibitors (ICIs), that show high efficacy in patients with advanced HCC. Along with the high efficacy of novel drugs, DILI has become a pivotal issue in the use of new drugs, including ICIs. This review demonstrates the immunological mechanism of DILI, including the innate and adaptive immune systems. Moreover, it aims to provide drug treatment targets, describe the mechanisms of DILI, and detail the management of DILI caused by drugs for HCC and LT.
Collapse
Affiliation(s)
- Soon Kyu Lee
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun Sun Jung
- Department of Pathology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jung Hyun Kwon
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
44
|
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: 32] [Impact Index Per Article: 16.0] [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
|
45
|
Iyoda T, Yamasaki S, Ueda S, Shimizu K, Fujii SI. Natural Killer T and Natural Killer Cell-Based Immunotherapy Strategies Targeting Cancer. Biomolecules 2023; 13:biom13020348. [PMID: 36830717 PMCID: PMC9953375 DOI: 10.3390/biom13020348] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Both natural killer T (NKT) and natural killer (NK) cells are innate cytotoxic lymphoid cells that produce inflammatory cytokines and chemokines, and their role in the innate immune response to tumors and microorganisms has been investigated. Especially, emerging evidence has revealed their status and function in the tumor microenvironment (TME) of tumor cells. Some bacteria producing NKT cell ligands have been identified to exert antitumor effects, even in the TME. By contrast, tumor-derived lipids or metabolites may reportedly suppress NKT and NK cells in situ. Since NKT and NK cells recognize stress-inducible molecules or inhibitory molecules on cancer cells, their status or function depends on the balance between inhibitory and activating receptor signals. As a recent strategy in cancer immunotherapy, the mobilization or restoration of endogenous NKT or NK cells by novel vaccines or therapies has become a focus of research. As a new biological evidence, after activation, effector memory-type NKT cells lasted in tumor-bearing models, and NK cell-based immune checkpoint inhibition potentiated the enhancement of NK cell cytotoxicity against cancer cells in preclinical and clinical trials. Furthermore, several new modalities based on the characteristics of NKT and NK cells, including artificial adjuvant vector cells, chimeric antigen receptor-expressing NK or NKT cell therapy, or their combination with immune checkpoint blockade have been developed. This review examines challenges and future directions for improving these therapies.
Collapse
Affiliation(s)
- Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Shogo Ueda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
- RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama 230-0045, Japan
- Correspondence: (K.S.); (S.F.); Tel.:+ 81-45-503-7062 (S.F.); Fax: +81-45-503-7061 (S.F.)
| | - Shin-ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
- RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama 230-0045, Japan
- Correspondence: (K.S.); (S.F.); Tel.:+ 81-45-503-7062 (S.F.); Fax: +81-45-503-7061 (S.F.)
| |
Collapse
|
46
|
Spencer Clinton JL, Vogt MB, Kneubehl AR, Hibl BM, Paust S, Rico-Hesse R. Sialokinin in mosquito saliva shifts human immune responses towards intracellular pathogens. PLoS Negl Trop Dis 2023; 17:e0011095. [PMID: 36735632 PMCID: PMC9897557 DOI: 10.1371/journal.pntd.0011095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Mosquito saliva is a mix of numerous proteins that are injected into the skin while the mosquito searches for a blood meal. While mosquito saliva is known to be immunogenic, the salivary components driving these immune responses, as well as the types of immune responses that occur, are not well characterized. We investigated the effects of one potential immunomodulatory mosquito saliva protein, sialokinin, on the human immune response. We used flow cytometry to compare human immune cell populations between humanized mice bitten by sialokinin knockout mosquitoes or injected with sialokinin, and compared them to those bitten by wild-type mosquitoes, unbitten, or saline-injected control mice. Humanized mice received 4 mosquito bites or a single injection, were euthanized after 7 days, and skin, spleen, bone marrow, and blood were harvested for immune cell profiling. Our results show that bites from sialokinin knockout mosquitoes induced monocyte and macrophage populations in the skin, blood, bone marrow, and spleens, and primarily affected CD11c- cell populations. Other increased immune cells included plasmacytoid dendritic cells in the blood, natural killer cells in the skin and blood, and CD4+ T cells in all samples analyzed. Conversely, we observed that mice bitten with sialokinin knockout mosquitoes had decreased NKT cell populations in the skin, and fewer B cells in the blood, spleen, and bone marrow. Taken together, we demonstrated that sialokinin knockout saliva induces elements of a TH1 cellular immune response, suggesting that the sialokinin peptide is inducing a TH2 cellular immune response during wild-type mosquito biting. These findings are an important step towards understanding how mosquito saliva modulates the human immune system and which components of saliva may be critical for arboviral infection. By identifying immunomodulatory salivary proteins, such as sialokinin, we can develop vaccines against mosquito saliva components and direct efforts towards blocking arboviral infections.
Collapse
Affiliation(s)
- Jennifer L. Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Megan B. Vogt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alexander R. Kneubehl
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Brianne M. Hibl
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Silke Paust
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California, United States of America
| | - Rebecca Rico-Hesse
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
47
|
Antitumor Immunity Exerted by Natural Killer and Natural Killer T Cells in the Liver. J Clin Med 2023; 12:jcm12030866. [PMID: 36769513 PMCID: PMC9917438 DOI: 10.3390/jcm12030866] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
The liver plays crucial roles in systemic immunity and greatly contributes to the systemic defense mechanism. Antitumor immunity in the liver is especially critical for the defense against systemic tumor cell dissemination. To achieve effective defense against metastatic tumor cells, liver immune cells with powerful cytotoxic activities construct a potent defense mechanism. In the liver, as compared with other organs, there is a significantly more intense percentage of innate immune lymphocytes, such as natural killer (NK) and NKT cells. These characteristic lymphocytes survey the portal blood transferred to the liver from the alimentary tract and eliminate malignant cells with their robust cytotoxic ability. Additionally, with their active cytokine-producing capacity, these innate lymphocytes initiate immunological sequences by adaptive immune cells. Therefore, they are crucial contributors to systemic antitumor immunity. These attractive immune cells help conduct a fundamental investigation of tumor immunity and act as a target of clinical measures for cancer therapies. This review discusses the mechanisms of these innate lymphocytes regarding recognition and cytotoxicity against tumor cells and the possibility of clinical applications for therapeutic measures.
Collapse
|
48
|
Kratzmeier C, Singh S, Asiedu EB, Webb TJ. Current Developments in the Preclinical and Clinical use of Natural Killer T cells. BioDrugs 2023; 37:57-71. [PMID: 36525216 PMCID: PMC9756707 DOI: 10.1007/s40259-022-00572-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2022] [Indexed: 12/23/2022]
Abstract
Natural killer T (NKT) cells play a pivotal role as a bridge between the innate and the adaptive immune response and are instrumental in the regulation of homeostasis. In this review, we discuss the potential for NKT cells to serve as biodrugs in viral infections and in cancer. NKT cells are being investigated for their use as a prognostic biomarker, an immune adjuvant, and as a form of cellular therapy. Historically, the clinical utility of NKT cells was hampered by their low frequency in the blood, discrepancies in nomenclature, and challenges with ex vivo expansion. However, recent advances in the field have permitted the development of several NKT cell-based preclinical and clinical strategies. These new developments pave the way for the successful implementation of NKT cell-based approaches for the treatment of human disease.
Collapse
Affiliation(s)
- Christina Kratzmeier
- Department of Microbiology and Immunology, and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 685 West Baltimore St, HSF I-Room 380, Baltimore, MD, 21201, USA
| | - Sasha Singh
- Department of Microbiology and Immunology, and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 685 West Baltimore St, HSF I-Room 380, Baltimore, MD, 21201, USA
| | - Emmanuel B Asiedu
- Department of Microbiology and Immunology, and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 685 West Baltimore St, HSF I-Room 380, Baltimore, MD, 21201, USA
| | - Tonya J Webb
- Department of Microbiology and Immunology, and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 685 West Baltimore St, HSF I-Room 380, Baltimore, MD, 21201, USA.
| |
Collapse
|
49
|
Comparison of the Single-Cell Immune Landscape of Testudines from Different Habitats. Cells 2022; 11:cells11244023. [PMID: 36552787 PMCID: PMC9816942 DOI: 10.3390/cells11244023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Testudines, also known as living fossils, have evolved diversely and comprise many species that occupy a variety of ecological niches. However, the immune adaptation of testudines to the different ecological niches remains poorly understood. This study compared the composition, function, and differentiation trajectories of peripheral immune cells in testudines (Chelonia mydas, Trachemys scripta elegans, Chelonoidis carbonaria, and Pelodiscus sinensis) from different habitats using the single-cell RNA sequencing (scRNA-seq) technique. The results showed that T. scripta elegans, which inhabits freshwater and brackish environments, had the most complex composition of peripheral immune cells, with 11 distinct immune cell subsets identified in total. The sea turtle C. mydas, had the simplest composition of peripheral immune cells, with only 5 distinct immune cell clusters. Surprisingly, neither basophils were found in C. mydas nor T cells in C. carbonaria. Basophil subsets in peripheral blood were identified for the first time; two basophil subtypes (GATA2-high-basophils and GATA2-low-basophils) were observed in the peripheral blood of T. scripta elegans. In addition, ACKR4 cells, CD4 T cells, CD7 T cells, serotriflin cells, and ficolin cells were specifically identified in the peripheral blood of T. scripta elegans. Furthermore, LY6G6C cells, SPC24 cells, and NKT cells were specifically observed in C. carbonaria. Moreover, there were differences in the functional status and developmental trajectory of peripheral immune cells among the testudine species. The identification of specific features of peripheral immune cells in testudines from different habitats may enable elucidation of the adaptation mechanism of testudines to various ecological niches.
Collapse
|
50
|
Li YR, Zeng S, Dunn ZS, Zhou Y, Li Z, Yu J, Wang YC, Ku J, Cook N, Kramer A, Yang L. Off-the-shelf third-party HSC-engineered iNKT cells for ameliorating GvHD while preserving GvL effect in the treatment of blood cancers. iScience 2022; 25:104859. [PMID: 36034226 PMCID: PMC9399487 DOI: 10.1016/j.isci.2022.104859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
Allo-HSCT is a curative therapy for hematologic malignancies owing to GvL effect mediated by alloreactive T cells; however, the same T cells also mediate GvHD, a severe side effect limiting the widespread application of allo-HSCT in clinics. Invariant natural killer T (iNKT) cells can ameliorate GvHD while preserving GvL effect, but the clinical application of these cells is restricted by their scarcity. Here, we report the successful generation of third-party HSC-engineered human iNKT (3rdHSC-iNKT) cells using a method combining HSC gene engineering and in vitro HSC differentiation. The 3rdHSC-iNKT cells closely resembled the CD4-CD8-/+ subsets of endogenous human iNKT cells in phenotype and functionality. These cells displayed potent anti-GvHD functions by eliminating antigen-presenting myeloid cells in vitro and in xenograft models without negatively impacting tumor eradication by allogeneic T cells in preclinical models of lymphoma and leukemia, supporting 3rdHSC-iNKT cells as a promising off-the-shelf cell therapy candidate for GvHD prophylaxis.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samuel Zeng
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zachary Spencer Dunn
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Yang Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhe Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yu-Chen Wang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Josh Ku
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Noah Cook
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adam Kramer
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|