1
|
Pellicci DG, Tavakolinia N, Perriman L, Berzins SP, Menne C. Thymic development of human natural killer T cells: recent advances and implications for immunotherapy. Front Immunol 2024; 15:1441634. [PMID: 39267746 PMCID: PMC11390520 DOI: 10.3389/fimmu.2024.1441634] [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: 05/31/2024] [Accepted: 08/05/2024] [Indexed: 09/15/2024] Open
Abstract
Invariant natural killer T (iNKT) cells are a subset of lipid-reactive, unconventional T cells that have anti-tumor properties that make them a promising target for cancer immunotherapy. Recent studies have deciphered the developmental pathway of human MAIT and Vγ9Vδ2 γδ-T cells as well as murine iNKT cells, yet our understanding of human NKT cell development is limited. Here, we provide an update in our understanding of how NKT cells develop in the human body and how knowledge regarding their development could enhance human treatments by targeting these cells.
Collapse
Affiliation(s)
- Daniel G Pellicci
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Naeimeh Tavakolinia
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, Australia
| | - Louis Perriman
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Fiona Elsey Cancer Institute, Ballarat, VIC, Australia
- Federation University Australia, Ballarat, VIC, Australia
| | - Stuart P Berzins
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, Australia
- Federation University Australia, Ballarat, VIC, Australia
| | | |
Collapse
|
2
|
Pagliuca S, Schmid C, Santoro N, Simonetta F, Battipaglia G, Guillaume T, Greco R, Onida F, Sánchez-Ortega I, Yakoub-Agha I, Kuball J, Hazenberg MD, Ruggeri A. Donor lymphocyte infusion after allogeneic haematopoietic cell transplantation for haematological malignancies: basic considerations and best practice recommendations from the EBMT. Lancet Haematol 2024; 11:e448-e458. [PMID: 38796194 DOI: 10.1016/s2352-3026(24)00098-x] [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: 02/14/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 05/28/2024]
Abstract
Since the early description of three patients with relapsed leukaemia after allogeneic haematopoietic cell transplantation (HCT) who obtained complete remission after donor lymphocyte infusions (DLIs), the added value of this procedure to induce or maintain graft-versus-leukaemia immunity has been undisputed. For more than 30 years, DLIs have become common practice as prophylactic, pre-emptive, or therapeutic immunotherapy. However, as with many aspects of allogeneic HCT, centres have developed their own routines and practices, and many questions related to the optimal applications and toxicity, or to the immunobiology of DLI induced tumour-immunity, remain. As a part of the Practice Harmonization and Guidelines Committee and the Cellular Therapy and Immunobiology Working Party of the European Society for Blood and Marrow Transplantation effort, a panel of experts with clinical and translational knowledge in transplantation immunology and cellular therapy met during a 2-day workshop in September, 2023, in Lille, France, and developed a set of consensus-based recommendations for the application of unmanipulated DLI after allogeneic HCT for haematological malignancies. Given the absence of prospective data in the majority of publications, these recommendations are mostly based on retrospective studies and expert consensus.
Collapse
Affiliation(s)
- Simona Pagliuca
- Department of Hematology, Nancy University Hospital, Nancy, France; UMR 7365, IMoPA, Lorraine University, CNRS, Vandœuvre-lès-Nancy, France
| | - Christoph Schmid
- Department of Haematology and Oncology, Augsburg University Hospital and Medical Faculty Comprehensive Cancer Center, Bavarian Cancer Research Center, Augsburg, Germany
| | - Nicole Santoro
- Haematology Unit, Department of Oncology and Hematology, Santo Spirito Hospital, Pescara, Italy
| | - Federico Simonetta
- Division of Haematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | - Giorgia Battipaglia
- Haematology Department and Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Thierry Guillaume
- Division of Haematology, Nantes University Hospital, Nantes, France; INSERM U1232 CNRS, CRCINA, Nantes, France
| | - Raffaella Greco
- Haematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Onida
- Haematology and BMT Unit, ASST Fatebenefratelli Sacco, University of Milan, Milan, Italy
| | | | | | - Jurgen Kuball
- Department of Haematology and Center for Translational Immunology, UMC Utrecht, Utrecht, Netherlands
| | - Mette D Hazenberg
- Department of Haematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Annalisa Ruggeri
- Haematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
3
|
Maurer K, Antin JH. The graft versus leukemia effect: donor lymphocyte infusions and cellular therapy. Front Immunol 2024; 15:1328858. [PMID: 38558819 PMCID: PMC10978651 DOI: 10.3389/fimmu.2024.1328858] [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: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many hematologic malignancies as well as non-malignant conditions. Part of the curative basis underlying HSCT for hematologic malignancies relies upon induction of the graft versus leukemia (GVL) effect in which donor immune cells recognize and eliminate residual malignant cells within the recipient, thereby maintaining remission. GVL is a clinically evident phenomenon; however, specific cell types responsible for inducing this effect and molecular mechanisms involved remain largely undefined. One of the best examples of GVL is observed after donor lymphocyte infusions (DLI), an established therapy for relapsed disease or incipient/anticipated relapse. DLI involves infusion of peripheral blood lymphocytes from the original HSCT donor into the recipient. Sustained remission can be observed in 20-80% of patients treated with DLI depending upon the underlying disease and the intrinsic burden of targeted cells. In this review, we will discuss current knowledge about mechanisms of GVL after DLI, experimental strategies for augmenting GVL by manipulation of DLI (e.g. neoantigen vaccination, specific cell type selection/depletion) and research outlook for improving DLI and cellular immunotherapies for hematologic malignancies through better molecular definition of the GVL effect.
Collapse
Affiliation(s)
| | - Joseph H. Antin
- Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Lv Z, Luo F, Chu Y. Strategies for overcoming bottlenecks in allogeneic CAR-T cell therapy. Front Immunol 2023; 14:1199145. [PMID: 37554322 PMCID: PMC10405079 DOI: 10.3389/fimmu.2023.1199145] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Patient-derived autologous chimeric antigen receptor (CAR)-T cell therapy is a revolutionary breakthrough in immunotherapy and has made impressive progress in both preclinical and clinical studies. However, autologous CAR-T cells still have notable drawbacks in clinical manufacture, such as long production time, variable cell potency and possible manufacturing failures. Allogeneic CAR-T cell therapy is significantly superior to autologous CAR-T cell therapy in these aspects. The use of allogeneic CAR-T cell therapy may provide simplified manufacturing process and allow the creation of 'off-the-shelf' products, facilitating the treatments of various types of tumors at less delivery time. Nevertheless, severe graft-versus-host disease (GvHD) or host-mediated allorejection may occur in the allogeneic setting, implying that addressing these two critical issues is urgent for the clinical application of allogeneic CAR-T cell therapy. In this review, we summarize the current approaches to overcome GvHD and host rejection, which empower allogeneic CAR-T cell therapy with a broader future.
Collapse
Affiliation(s)
- Zixin Lv
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, China
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Biotherapy Research Center, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Stavrou V, Fultang L, Booth S, De Simone D, Bartnik A, Scarpa U, Gneo L, Panetti S, Potluri S, Almowaled M, Barlow J, Jankevics A, Lloyd G, Southam A, Priestman DA, Cheng P, Dunn W, Platt F, Endou H, Craddock C, Keeshan K, Mussai F, De Santo C. Invariant NKT cells metabolically adapt to the acute myeloid leukaemia environment. Cancer Immunol Immunother 2023; 72:543-560. [PMID: 35962843 PMCID: PMC9947083 DOI: 10.1007/s00262-022-03268-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/27/2022] [Indexed: 10/15/2022]
Abstract
Acute myeloid leukaemia (AML) creates an immunosuppressive environment to conventional T cells through Arginase 2 (ARG2)-induced arginine depletion. We identify that AML blasts release the acute phase protein serum amyloid A (SAA), which acts in an autocrine manner to upregulate ARG2 expression and activity, and promote AML blast viability. Following in vitro cross-talk invariant natural killer T (iNKT) cells become activated, upregulate mitochondrial capacity, and release IFN-γ. iNKT retain their ability to proliferate and be activated despite the low arginine AML environment, due to the upregulation of Large Neutral Amino Acid Transporter-1 (LAT-1) and Argininosuccinate Synthetase 1 (ASS)-dependent amino acid pathways, resulting in AML cell death. T cell proliferation is restored in vitro and in vivo. The capacity of iNKT cells to restore antigen-specific T cell immunity was similarly demonstrated against myeloid-derived suppressor cells (MDSCs) in wild-type and Jα18-/- syngeneic lymphoma-bearing models in vivo. Thus, stimulation of iNKT cell activity has the potential as an immunotherapy against AML or as an adjunct to boost antigen-specific T cell immunotherapies in haematological or solid cancers.
Collapse
Affiliation(s)
- Victoria Stavrou
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Livingstone Fultang
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Sarah Booth
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Daniele De Simone
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Arekdiusz Bartnik
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ugo Scarpa
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Luciana Gneo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Silvia Panetti
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Sandeep Potluri
- Institute of Cancer and Genomics, University of Birmingham, Birmingham, B15 2TT, UK
| | - Meaad Almowaled
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, G12 0YN, UK
| | - Jonathan Barlow
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Andris Jankevics
- School of Biosciences and Phenome Centre Birmingham, University of Birmingham, Birmingham, B15 2TT, UK
| | - Gavin Lloyd
- School of Biosciences and Phenome Centre Birmingham, University of Birmingham, Birmingham, B15 2TT, UK
| | - Andrew Southam
- School of Biosciences and Phenome Centre Birmingham, University of Birmingham, Birmingham, B15 2TT, UK
| | - David A Priestman
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Paul Cheng
- Bio-Cancer Treatment International, Hong Kong Science Park, Hong Kong, China
| | - Warwick Dunn
- Institute of Cancer and Genomics, University of Birmingham, Birmingham, B15 2TT, UK.,Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Frances Platt
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Hitoshi Endou
- J-Pharma Co. Ltd, Yokohama, Kanagawa, 230-0046, Japan
| | - Charles Craddock
- Institute of Cancer and Genomics, University of Birmingham, Birmingham, B15 2TT, UK
| | - Karen Keeshan
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, G12 0YN, UK
| | - Francis Mussai
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Carmela De Santo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
6
|
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: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
7
|
Odak I, Sikora R, Riemann L, Bayir LM, Beck M, Drenker M, Xiao Y, Schneider J, Dammann E, Stadler M, Eder M, Ganser A, Förster R, Koenecke C, Schultze-Florey CR. Spectral flow cytometry cluster analysis of therapeutic donor lymphocyte infusions identifies T cell subsets associated with outcome in patients with AML relapse. Front Immunol 2022; 13:999163. [PMID: 36275657 PMCID: PMC9579313 DOI: 10.3389/fimmu.2022.999163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Identification of immune phenotypes linked to durable graft-versus-leukemia (GVL) response following donor lymphocyte infusions (DLI) is of high clinical relevance. In this prospective observational study of 13 AML relapse patients receiving therapeutic DLI, we longitudinally investigated changes in differentiation stages and exhaustion markers of T cell subsets using cluster analysis of 30-color spectral flow cytometry during 24 months follow-up. DLI cell products and patient samples after DLI were analyzed and correlated to the clinical outcome. Analysis of DLI cell products revealed heterogeneity in the proportions of naïve and antigen experienced T cells. Cell products containing lower levels of effector memory (eff/m) cells and higher amounts of naïve CD4+ and CD8+ T cells were associated with long-term remission. Furthermore, investigation of patient blood samples early after DLI showed that patients relapsing during the study period, had higher levels of CD4+ eff/m T cells and expressed a mosaic of surface molecules implying an exhausted functional state. Of note, this observation preceded the clinical diagnosis of relapse by five months. On the other hand, patients with continuous remission retained lower levels of exhausted CD4+ eff/m T cells more than four months post DLI. Moreover, lower frequencies of exhausted CD8+ eff/m T cells as well as higher amounts of CD4+temra CD45RO+ T cells were present in this group. These results imply the formation of functional long-term memory pool of T cells. Finally, unbiased sample analysis showed that DLI cell products with low levels of eff/m cells both in CD4+ and CD8+ T cell subpopulations associate with a lower relapse incidence. Additionally, competing risk analysis of patient samples taken early after DLI revealed that patients with high amounts of exhausted CD4+ eff/m T cells in their blood exhibited significantly higher rates of relapse. In conclusion, differentially activated T cell clusters, both in the DLI product and in patients post infusion, were associated with AML relapse after DLI. Our study suggests that differences in DLI cell product composition might influence GVL. In-depth monitoring of T cell dynamics post DLI might increase safety and efficacy of this immunotherapy, while further studies are needed to assess the functionality of T cells found in the DLI.
Collapse
Affiliation(s)
- Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- *Correspondence: Christian R. Schultze-Florey, ; Ivan Odak,
| | - Ruth Sikora
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Lennart Riemann
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Lâle M. Bayir
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Maleen Beck
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Melanie Drenker
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Yankai Xiao
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Jessica Schneider
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Elke Dammann
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Michael Stadler
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Matthias Eder
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christian Koenecke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Christian R. Schultze-Florey
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- *Correspondence: Christian R. Schultze-Florey, ; Ivan Odak,
| |
Collapse
|
8
|
Hess NJ, S Bharadwaj N, Bobeck EA, McDougal CE, Ma S, Sauer JD, Hudson AW, Gumperz JE. iNKT cells coordinate immune pathways to enable engraftment in nonconditioned hosts. Life Sci Alliance 2021; 4:e202000999. [PMID: 34112724 PMCID: PMC8200291 DOI: 10.26508/lsa.202000999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/05/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a conserved population of innate T lymphocytes that interact with key antigen-presenting cells to modulate adaptive T-cell responses in ways that can either promote protective immunity, or limit pathological immune activation. Understanding the immunological networks engaged by iNKT cells to mediate these opposing functions is a key pre-requisite to effectively using iNKT cells for therapeutic applications. Using a human umbilical cord blood xenotransplantation model, we show here that co-transplanted allogeneic CD4+ iNKT cells interact with monocytes and T cells in the graft to coordinate pro-hematopoietic and immunoregulatory pathways. The nexus of iNKT cells, monocytes, and cord blood T cells led to the release of cytokines (IL-3, GM-CSF) that enhance hematopoietic stem and progenitor cell activity, and concurrently induced PGE2-mediated suppression of T-cell inflammatory responses that limit hematopoietic stem and progenitor cell engraftment. This resulted in successful long-term hematopoietic engraftment without pretransplant conditioning, including multi-lineage human chimerism and colonization of the spleen by antibody-producing human B cells. These results highlight the potential for using iNKT cellular immunotherapy to improve rates of hematopoietic engraftment independently of pretransplant conditioning.
Collapse
Affiliation(s)
- Nicholas J Hess
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nikhila S Bharadwaj
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Elizabeth A Bobeck
- Department of Animal Science, 201F Kildee Hall, Iowa State University, Ames, IA, USA
| | - Courtney E McDougal
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Shidong Ma
- QLB Biotherapeutics, Inc., Boston, MA, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Amy W Hudson
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jenny E Gumperz
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
9
|
Andrlová H, van den Brink MRM, Markey KA. An Unconventional View of T Cell Reconstitution After Allogeneic Hematopoietic Cell Transplantation. Front Oncol 2021; 10:608923. [PMID: 33680931 PMCID: PMC7930482 DOI: 10.3389/fonc.2020.608923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/31/2020] [Indexed: 01/02/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is performed as curative-intent therapy for hematologic malignancies and non-malignant hematologic, immunological and metabolic disorders, however, its broader implementation is limited by high rates of transplantation-related complications and a 2-year mortality that approaches 50%. Robust reconstitution of a functioning innate and adaptive immune system is a critical contributor to good long-term patient outcomes, primarily to prevent and overcome post-transplantation infectious complications and ensure adequate graft-versus-leukemia effects. There is increasing evidence that unconventional T cells may have an important immunomodulatory role after allo-HCT, which may be at least partially dependent on the post-transplantation intestinal microbiome. Here we discuss the role of immune reconstitution in allo-HCT outcome, focusing on unconventional T cells, specifically mucosal-associated invariant T (MAIT) cells, γδ (gd) T cells, and invariant NK T (iNKT) cells. We provide an overview of the mechanistic preclinical and associative clinical studies that have been performed. We also discuss the emerging role of the intestinal microbiome with regard to hematopoietic function and overall immune reconstitution.
Collapse
Affiliation(s)
- Hana Andrlová
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Marcel R. M. van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Division of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Kate A. Markey
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Division of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
10
|
[Invariant natural killer T cells : Cellular immune regulators with cytotoxic potential]. DER PATHOLOGE 2021; 41:134-137. [PMID: 33156364 DOI: 10.1007/s00292-020-00829-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Invariant natural killer T cells (iNKT cells) are a small subset of T lymphocytes that are equipped with various immunoregulatory cytokines and cytotoxic effector molecules. Immune responses can be modulated efficiently by their interaction with other cells of the innate and adaptive immune system. Also, iNKT cells can promote apoptosis of malignant cells. Therefore, iNKT cells play a particular role in infectious diseases, malignant diseases, autoimmunity and alloimmunity. After allogeneic hematopoietic cell transplantation, iNKT cells induce immune tolerance and promote graft-versus-tumor effects. Recent advances in automated cell processing under good manufacturing practice (GMP) conditions and genetic modifications of effector cells pave the way for clinical translation of iNKT cell therapy.
Collapse
|
11
|
Schmid H, Ribeiro EM, Secker KA, Duerr-Stoerzer S, Keppeler H, Dong R, Munz T, Schulze-Osthoff K, Hailfinger S, Schneidawind C, Schneidawind D. Human invariant natural killer T cells promote tolerance by preferential apoptosis induction of conventional dendritic cells. Haematologica 2021; 107:427-436. [PMID: 33440919 PMCID: PMC8804566 DOI: 10.3324/haematol.2020.267583] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Indexed: 11/20/2022] Open
Abstract
Graft-versus-host disease (GvHD) is a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation. We recently showed in murine studies and in vitro human models that adoptively transferred invariant natural killer T (iNKT) cells protect from GvHD and promote graft-versus-leukemia effects. The cellular mechanisms underlying GvHD prevention by iNKT cells in humans, however, remain unknown. In order to study relevant cellular interactions, dendritic cells (DC) were either generated from monocytes or isolated directly from blood of healthy donors or GvHD patients and co-cultured in a mixed lymphocyte reaction (MLR) with T cells obtained from healthy donors or transplantation bags. Addition of culture-expanded iNKT cells to the MLR-induced DC apoptosis in a cell contact-dependent manner, thereby preventing T-cell activation and proliferation. Annexin V/propidium iodide staining and image stream assays showed that CD4+CD8–, CD4–CD8+ and double negative iNKT cells are similarly able to induce DC apoptosis. Further MLR assays revealed that conventional DC (cDC) but not plasmacytoid DC (pDC) could induce alloreactive T-cell activation and proliferation. Interestingly, cDC were also more susceptible to apoptosis induced by iNKT cells, which correlates with their higher CD1d expression, leading to a bias in favor of pDC. Remarkably, these results could also be observed in GvHD patients. We propose a new mechanism how ex vivo expanded human iNKT cells prevent alloreactivity of T cells. iNKT cells modulate T-cell responses by selective apoptosis of DC subsets, resulting in suppression of T-cell activation and proliferation while enabling beneficial immune responses through pDC.
Collapse
Affiliation(s)
- Hannes Schmid
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Emmanuelle M Ribeiro
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Kathy-Ann Secker
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Silke Duerr-Stoerzer
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Hildegard Keppeler
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Ruoyun Dong
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Timo Munz
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | | | - Stephan Hailfinger
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tuebingen
| | - Corina Schneidawind
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Dominik Schneidawind
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen.
| |
Collapse
|
12
|
Khaldoyanidi S, Nagorsen D, Stein A, Ossenkoppele G, Subklewe M. Immune Biology of Acute Myeloid Leukemia: Implications for Immunotherapy. J Clin Oncol 2021; 39:419-432. [PMID: 33434043 PMCID: PMC8078464 DOI: 10.1200/jco.20.00475] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
| | | | - Anthony Stein
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Gerrit Ossenkoppele
- Amsterdam University Medical Center, Location VU University Medical Center, Amsterdam, the Netherlands
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
13
|
Li Y, Su R, Chen J. Co-culture Systems of Drug-Treated Acute Myeloid Leukemia Cells and T Cells for In Vitro and In Vivo Study. STAR Protoc 2020; 1. [PMID: 32995754 PMCID: PMC7521668 DOI: 10.1016/j.xpro.2020.100097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A combination of immunotherapy and chemotherapy strategies could strengthen antitumor effects. This protocol elucidates a robust method via co-culturing drug pre-treated acute myeloid leukemia cells with CD3+ T cells, derived from leukoreduction system chambers, for in vitro and in vivo study. We optimized several aspects of the procedures, including timing of drug treatment, quantification of tumor cells, and approach of combination of CD3+ T cells with drug treatment in vivo. This enables the readouts of the interplay between drugs and T cells. For complete details on the use and execution of this protocol, please refer to Su et al. (2020). Generate CD3+ T cells from leukoreduction system chambers using magnetic separation Co-culture T cells with drug-pretreated fluorescently labeled tumor cells Determine T cell toxicity in the co-culture system via absolute counting beads Combine T cells and drug treatment in the xenograft mouse with luciferase-labeled AML cells
Collapse
Affiliation(s)
- Yangchan Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA.,Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China.,These authors contributed equally
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA.,These authors contributed equally.,Technical Contact
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA.,Lead Contact
| |
Collapse
|