1
|
Ramos TL, Bolivar-Wagers S, Jin S, Thangavelu G, Simonetta F, Lin PY, Hirai T, Saha A, Koehn B, Su LL, Picton LK, Baker J, Lohmeyer JK, Riddle M, Eide C, Tolar J, Panoskaltsis-Mortari A, Wagner JE, Garcia KC, Negrin RS, Blazar BR. Prevention of acute GVHD using an orthogonal IL-2/IL-2Rβ system to selectively expand regulatory T cells in vivo. Blood 2023; 141:1337-1352. [PMID: 36564052 PMCID: PMC10082364 DOI: 10.1182/blood.2022018440] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/09/2022] [Accepted: 12/01/2022] [Indexed: 12/25/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative option for patients with hematological disorders and bone marrow (BM) failure syndromes. Graft-versus-host disease (GVHD) remains a leading cause of morbidity posttransplant. Regulatory T cell (Treg) therapies are efficacious in ameliorating GVHD but limited by variable suppressive capacities and the need for a high therapeutic dose. Here, we sought to expand Treg in vivo by expressing an orthogonal interleukin 2 receptor β (oIL-2Rβ) that would selectively interact with oIL-2 cytokine and not wild-type (WT) IL-2. To test whether the orthogonal system would preferentially drive donor Treg expansion, we used a murine major histocompatibility complex-disparate GVHD model of lethally irradiated BALB/c mice given T cell-depleted BM from C57BL/6 (B6) mice alone or together with B6Foxp3+GFP+ Treg or oIL-2Rβ-transduced Treg at low cell numbers that typically do not control GVHD with WT Treg. On day 2, B6 activated T cells (Tcons) were injected to induce GVHD. Recipients were treated with phosphate-buffered saline (PBS) or oIL-2 daily for 14 days, then 3 times weekly for an additional 14 days. Mice treated with oIL-2Rβ Treg and oIL-2 compared with those treated with PBS had enhanced GVHD survival, in vivo selective expansion of Tregs, and greater suppression of Tcon expansion in secondary lymphoid organs and intestines. Importantly, oIL-2Rβ Treg maintained graft-versus-tumor (GVT) responses in 2 distinct tumor models (A20 and MLL-AF9). These data demonstrate a novel approach to enhance the efficacy of Treg therapy in allo-HSCT using an oIL-2/oIL-2Rβ system that allows for selective in vivo expansion of Treg leading to GVHD protection and GVT maintenance.
Collapse
Affiliation(s)
- Teresa L. Ramos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA
| | - Sara Bolivar-Wagers
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Sujeong Jin
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Govindarajan Thangavelu
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Federico Simonetta
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA
- Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Po-Yu Lin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA
| | - Toshihito Hirai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA
- Department of Urology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Asim Saha
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Brent Koehn
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Leon L. Su
- Department of Molecular and Cellular Physiology, Department of Structural Biology, School of Medicine, Stanford University, Stanford, CA
| | - Lora K. Picton
- Department of Molecular and Cellular Physiology, Department of Structural Biology, School of Medicine, Stanford University, Stanford, CA
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA
| | - Juliane K. Lohmeyer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA
| | - Megan Riddle
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Cindy Eide
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Jakub Tolar
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - John E. Wagner
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - K. Christopher Garcia
- Department of Molecular and Cellular Physiology, Department of Structural Biology, School of Medicine, Stanford University, Stanford, CA
| | - Robert S. Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| |
Collapse
|
2
|
Ritacco C, Köse MC, Courtois J, Canti L, Beguin C, Dubois S, Vandenhove B, Servais S, Caers J, Beguin Y, Ehx G, Baron F. Post-transplant cyclophosphamide prevents xenogeneic graft-versus-host disease while depleting proliferating regulatory T cells. iScience 2023; 26:106085. [PMID: 36843851 PMCID: PMC9947306 DOI: 10.1016/j.isci.2023.106085] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/12/2022] [Accepted: 01/25/2023] [Indexed: 02/02/2023] Open
Abstract
Graft-versus-host disease (GVHD) remains a serious limitation of allogeneic hematopoietic cell transplantation (allo-HCT). While post-transplant administration of cyclophosphamide (PTCy) is increasingly used as GVHD prophylaxis, its precise mechanisms of action and its impact on graft-versus-leukemia effects have remained debated. Here, we studied the mechanisms of xenogeneic GVHD (xGVHD) prevention by PTCy in different humanized mouse models. We observed that PTCy attenuated xGVHD. Using flow cytometry and single-cell RNA-sequencing, we demonstrated that PTCy depleted proliferative CD8+ and conventional CD4+ T cells but also proliferative regulatory T cells (Treg). Further, T-cell receptor β variable region sequencing (TCRVB) analyses demonstrated that highly xenoreactive T-cell clones were depleted by PTCy. Although Treg frequencies were significantly higher in PTCy-treated than in control mice on day 21, xGVHD attenuation by PTCy was not abrogated by Treg depletion. Finally, we observed that PTCy did not abrogate graft-versus-leukemia effects.
Collapse
Affiliation(s)
- Caroline Ritacco
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium
| | - Murat Cem Köse
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium
| | - Justine Courtois
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium
| | - Lorenzo Canti
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium
| | - Charline Beguin
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium
| | - Sophie Dubois
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium
| | - Benoît Vandenhove
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium
| | - Sophie Servais
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium,Department of Medicine, Division of Hematology, CHU of Liège, Liège 4000, Belgium
| | - Jo Caers
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium,Department of Medicine, Division of Hematology, CHU of Liège, Liège 4000, Belgium
| | - Yves Beguin
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium,Department of Medicine, Division of Hematology, CHU of Liège, Liège 4000, Belgium
| | - Grégory Ehx
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium,Corresponding author
| | - Frédéric Baron
- Hematology Research Unit, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I³, University of Liège, Liège 4000, Belgium,Department of Medicine, Division of Hematology, CHU of Liège, Liège 4000, Belgium,Corresponding author
| |
Collapse
|
3
|
Using stroma-anchoring cytokines to augment ADCC: a phase 1 trial of F16IL2 and BI 836858 for posttransplant AML relapse. Blood Adv 2022; 6:3684-3696. [PMID: 35468621 DOI: 10.1182/bloodadvances.2021006909] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/14/2022] [Indexed: 11/20/2022] Open
Abstract
Natural killer (NK) cells are key effectors in cancer immunosurveillance and posttransplant immunity, but deficiency of environmental signals and insufficient tumor recognition may limit their activity. We hypothesized that the antibody-mediated anchoring of interleukin-2 (IL-2) to a spliced isoform of the extracellular matrix (ECM) glycoprotein tenascin-C would potentiate NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) against leukemic blasts. In this novel-novel combination, dose-escalation phase 1 trial, we enrolled patients with posttransplant acute myeloid leukemia (AML) relapse to evaluate the safety, pharmacokinetics, pharmacodynamics, and preliminary activity of the antibody-cytokine fusion F16IL2 (10-20x106 IU IV, days 1, 8, 15, and 22 of 28-day cycles) in combination with the anti-CD33 antibody BI 836858 (10-40 mg IV, 2 days after each F16IL2 infusion). Among 15 patients (median [range] age, 50 [20-68] years) treated across 4 dose levels (DL), 6 (40%) had received 2 or 3 prior transplantations. The most frequent adverse events were pyrexia, chills and infusion-related reactions, which were manageable, transient and of grade ≤ 2. One dose-limiting toxicity occurred at each of DL 3 (pulmonary edema) and 4 (GVHD). Three objective responses were observed among 7 patients treated at the 2 higher DL, whereas no responses occurred at the 2 starting DL. Combination therapy stimulated the expansion and activation of NK cells, including those expressing the FcγRIIIA/CD16 receptor. ECM-targeted IL-2 combined with anti-CD33 immunotherapy represents an innovative approach associated with acceptable safety and encouraging biologic and clinical activity in posttransplant AML relapse. This trial was registered at EudraCT (2015-004763-37).
Collapse
|
4
|
Nicholls J, Cao B, Le Texier L, Xiong LY, Hunter CR, Llanes G, Aguliar EG, Schroder WA, Phipps S, Lynch JP, Cao H, Heazlewood SY, Williams B, Clouston AD, Nefzger CM, Polo JM, Nilsson SK, Blazar BR, MacDonald KPA. Bone Marrow Regulatory T Cells Are a Unique Population, Supported by Niche-Specific Cytokines and Plasmacytoid Dendritic Cells, and Required for Chronic Graft-Versus-Host Disease Control. Front Cell Dev Biol 2021; 9:737880. [PMID: 34631716 PMCID: PMC8493124 DOI: 10.3389/fcell.2021.737880] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Regulatory T cell (Treg) reconstitution is essential for reestablishing tolerance and maintaining homeostasis following stem-cell transplantation. We previously reported that bone marrow (BM) is highly enriched in autophagy-dependent Treg and autophagy disruption leads to a significant Treg loss, particularly BM-Treg. To correct the known Treg deficiency observed in chronic graft-versus-host disease (cGVHD) patients, low dose IL-2 infusion has been administered, substantially increasing peripheral Treg (pTreg) numbers. However, as clinical responses were only seen in ∼50% of patients, we postulated that pTreg augmentation was more robust than for BM-Treg. We show that BM-Treg and pTreg have distinct characteristics, indicated by differential transcriptome expression for chemokine receptors, transcription factors, cell cycle control of replication and genes linked to Treg function. Further, BM-Treg were more quiescent, expressed lower FoxP3, were highly enriched for co-inhibitory markers and more profoundly depleted than splenic Treg in cGVHD mice. In vivo our data are consistent with the BM and not splenic microenvironment is, at least in part, driving this BM-Treg signature, as adoptively transferred splenic Treg that entered the BM niche acquired a BM-Treg phenotype. Analyses identified upregulated expression of IL-9R, IL-33R, and IL-7R in BM-Treg. Administration of the T cell produced cytokine IL-2 was required by splenic Treg expansion but had no impact on BM-Treg, whereas the converse was true for IL-9 administration. Plasmacytoid dendritic cells (pDCs) within the BM also may contribute to BM-Treg maintenance. Using pDC-specific BDCA2-DTR mice in which diptheria toxin administration results in global pDC depletion, we demonstrate that pDC depletion hampers BM, but not splenic, Treg homeostasis. Together, these data provide evidence that BM-Treg and splenic Treg are phenotypically and functionally distinct and influenced by niche-specific mediators that selectively support their respective Treg populations. The unique properties of BM-Treg should be considered for new therapies to reconstitute Treg and reestablish tolerance following SCT.
Collapse
Affiliation(s)
- Jemma Nicholls
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Benjamin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Laetitia Le Texier
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Laura Yan Xiong
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Christopher R. Hunter
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Genesis Llanes
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Ethan G. Aguliar
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Wayne A. Schroder
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Simon Phipps
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jason P. Lynch
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Huimin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Shen Y. Heazlewood
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Brenda Williams
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | | | - Christian M. Nefzger
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Jose M. Polo
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Susan K. Nilsson
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Kelli P. A. MacDonald
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
5
|
Hefazi M, Bolivar-Wagers S, Blazar BR. Regulatory T Cell Therapy of Graft-versus-Host Disease: Advances and Challenges. Int J Mol Sci 2021; 22:9676. [PMID: 34575843 PMCID: PMC8469916 DOI: 10.3390/ijms22189676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/03/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
Graft-versus-host disease (GVHD) is the leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Immunomodulation using regulatory T cells (Tregs) offers an exciting option to prevent and/or treat GVHD as these cells naturally function to maintain immune homeostasis, can induce tolerance following HSCT, and have a tissue reparative function. Studies to date have established a clinical safety profile for polyclonal Tregs. Functional enhancement through genetic engineering offers the possibility of improved potency, specificity, and persistence. In this review, we provide the most up to date preclinical and clinical data on Treg cell therapy with a particular focus on GVHD. We discuss the different Treg subtypes and highlight the pharmacological and genetic approaches under investigation to enhance the application of Tregs in allo-HSCT. Lastly, we discuss the remaining challenges for optimal clinical translation and provide insights as to future directions of the field.
Collapse
Affiliation(s)
- Mehrdad Hefazi
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Sara Bolivar-Wagers
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA;
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA;
| |
Collapse
|
6
|
Tkachev V, Kaminski J, Potter EL, Furlan SN, Yu A, Hunt DJ, McGuckin C, Zheng H, Colonna L, Gerdemann U, Carlson J, Hoffman M, Olvera J, English C, Baldessari A, Panoskaltsis-Mortari A, Watkins B, Qayed M, Suessmuth Y, Betz K, Bratrude B, Langston A, Horan JT, Ordovas-Montanes J, Shalek AK, Blazar BR, Roederer M, Kean LS. Spatiotemporal single-cell profiling reveals that invasive and tissue-resident memory donor CD8 + T cells drive gastrointestinal acute graft-versus-host disease. Sci Transl Med 2021; 13:13/576/eabc0227. [PMID: 33441422 DOI: 10.1126/scitranslmed.abc0227] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022]
Abstract
Organ infiltration by donor T cells is critical to the development of acute graft-versus-host disease (aGVHD) in recipients after allogeneic hematopoietic stem cell transplant (allo-HCT). However, deconvoluting the transcriptional programs of newly recruited donor T cells from those of tissue-resident T cells in aGVHD target organs remains a challenge. Here, we combined the serial intravascular staining technique with single-cell RNA sequencing to dissect the tightly connected processes by which donor T cells initially infiltrate tissues and then establish a pathogenic tissue residency program in a rhesus macaque allo-HCT model that develops aGVHD. Our results enabled creation of a spatiotemporal map of the transcriptional programs controlling donor CD8+ T cell infiltration into the primary aGVHD target organ, the gastrointestinal (GI) tract. We identified the large and small intestines as the only two sites demonstrating allo-specific, rather than lymphodepletion-driven, T cell infiltration. GI-infiltrating donor CD8+ T cells demonstrated a highly activated, cytotoxic phenotype while simultaneously developing a canonical tissue-resident memory T cell (TRM) transcriptional signature driven by interleukin-15 (IL-15)/IL-21 signaling. We found expression of a cluster of genes directly associated with tissue invasiveness, including those encoding adhesion molecules (ITGB2), specific chemokines (CCL3 and CCL4L1) and chemokine receptors (CD74), as well as multiple cytoskeletal proteins. This tissue invasion transcriptional signature was validated by its ability to discriminate the CD8+ T cell transcriptome of patients with GI aGVHD from those of GVHD-free patients. These results provide insights into the mechanisms controlling tissue occupancy of target organs by pathogenic donor CD8+ TRM cells during aGVHD in primate transplant recipients.
Collapse
Affiliation(s)
- Victor Tkachev
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - James Kaminski
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - E Lake Potter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20858, USA
| | - Scott N Furlan
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Alison Yu
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel J Hunt
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Connor McGuckin
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hengqi Zheng
- University of Washington, Seattle, WA 98195, USA
| | - Lucrezia Colonna
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Ulrike Gerdemann
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Michelle Hoffman
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Joe Olvera
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Chris English
- Washington National Primate Research Center, Seattle, WA 98195, USA
| | | | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55454, USA
| | | | - Muna Qayed
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Kayla Betz
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Brandi Bratrude
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - John T Horan
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jose Ordovas-Montanes
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Division of Gastroenterology, Boston Children's Hospital and Program in Immunology, Harvard Medical School, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Alex K Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55454, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20858, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Itacitinib prevents xenogeneic GVHD in humanized mice. Bone Marrow Transplant 2021; 56:2672-2681. [PMID: 34172892 DOI: 10.1038/s41409-021-01363-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/06/2021] [Accepted: 05/18/2021] [Indexed: 01/09/2023]
Abstract
We assessed the impact of the Janus Kinase (JAK) 1 inhibitor itacitinib on xenogeneic graft-versus-host disease (xGVHD). XGVHD was induced by i.v. injection 20 × 106 human peripheral blood mononuclear cells (hPBMC) in NSG mice on day 0. Itacitinib (3 mg, ≈120 mg/kg) or methylcellulose was administered by force-feeding twice a day from day 3 to day 28. Mice were followed for xGVHD score and survival. In addition, human T-cell engraftment and as well as human T-cell subtypes were monitored in blood on days 14, 21, and 28 after transplantation. We observed that itacitinib-treated mice had significantly longer survival than control mice (median 45 versus 33 days; P < 0.001). Further, they also had lower absolute numbers of human CD4+ T cells on days 21 and 28 after transplantation as well as of human CD8+ T cells on days 14, 21, and 28 after transplantation. In addition, itacitinib-treated mice had higher frequencies of human regulatory T cells (Treg) on days 21 and 28 after transplantation. In summary, our data indicate that itacitinib decreases human T-cell engraftment, increases Treg frequencies and attenuates xGVHD in NSG mice transplanted with hPBMC.
Collapse
|
8
|
Guil-Luna S, Sedlik C, Piaggio E. Humanized Mouse Models to Evaluate Cancer Immunotherapeutics. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2021. [DOI: 10.1146/annurev-cancerbio-050520-100526] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immunotherapy is at the forefront of cancer treatment. The advent of numerous novel approaches to cancer immunotherapy, including immune checkpoint antibodies, adoptive transfer of CAR (chimeric antigen receptor) T cells and TCR (T cell receptor) T cells, NK (natural killer) cells, T cell engagers, oncolytic viruses, and vaccines, is revolutionizing the treatment for different tumor types. Some are already in the clinic, and many others are underway. However, not all patients respond, resistance develops, and as available therapies multiply there is a need to further understand how they work, how to prioritize their clinical evaluation, and how to combine them. For this, animal models have been highly instrumental, and humanized mice models (i.e., immunodeficient mice engrafted with human immune and cancer cells) represent a step forward, although they have several limitations. Here, we review the different humanized models available today, the approaches to overcome their flaws, their use for the evaluation of cancer immunotherapies, and their anticipated evolution as tools to help personalized clinical decision-making.
Collapse
Affiliation(s)
- Silvia Guil-Luna
- Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain
| | - Christine Sedlik
- Translational Research Department, Institut Curie Research Center, INSERM U932, PSL Research University, 75248 Paris, France;,
| | - Eliane Piaggio
- Translational Research Department, Institut Curie Research Center, INSERM U932, PSL Research University, 75248 Paris, France;,
| |
Collapse
|
9
|
Kojima H, Kashiwakura Y, Kanno Y, Hashiguchi M, Kobata T. Fine-tuning of antigen-specific immune responses by regulatory T cells activated via antigen recognition-independent and humoral factor-dependent mechanisms. Scand J Immunol 2021; 93:e13020. [PMID: 33393095 DOI: 10.1111/sji.13020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 11/28/2022]
Abstract
CD4+ CD25+ Foxp3+ regulatory T cells (Tregs) are highly sensitive to IL-2, one of the many cytokines produced during immune responses, for their development, survival and functions. Although the effects of IL-2 administration on Tregs in vivo are well characterized, the effects on Tregs elicited by IL-2 produced during an immune response have not been elucidated. Hence, in this study, Treg behaviour during IL-2-producing immune responses was explored using in vivo and in vitro murine systems. The use of murine mixed lymphocyte culture (MLC) revealed that a large proportion of Tregs increased in size, accompanied by both cell death and proliferation status. Further, these large Tregs, which were found to not recognize specific antigens, were observed in MLCs as being functionally activated by various cytokines, including IL-2, produced by antigen-specific T cells. This 'bystander Treg activation' was also observed in mice with graft-versus-host reactions (GvHRs). Alternatively, effector cells from Treg-depleted MLCs exhibited lower antigen-specific responses or higher cross-reactivity as compared to control MLCs with Tregs. Taken together, these results suggest that Tregs are activated by cytokines, mainly IL-2, released from T cells that are activated by a specific antigen. Subsequently, these activated bystander Tregs contribute to the fine-tuning of highly antigen-specific immune responses.
Collapse
Affiliation(s)
- Hidefumi Kojima
- Department of Immunology, Dokkyo Medical University School of Medicine, Tochigi, Japan.,Division of Host Defense, Research Center for Advanced Medical Science, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Yuji Kashiwakura
- Department of Immunology, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Yumiko Kanno
- Department of Immunology, Dokkyo Medical University School of Medicine, Tochigi, Japan.,Division of Host Defense, Research Center for Advanced Medical Science, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Masaaki Hashiguchi
- Department of Immunology, Dokkyo Medical University School of Medicine, Tochigi, Japan.,Division of Host Defense, Research Center for Advanced Medical Science, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Tetsuji Kobata
- Department of Immunology, Dokkyo Medical University School of Medicine, Tochigi, Japan
| |
Collapse
|
10
|
Copsel S, Wolf D, Komanduri KV, Levy RB. The promise of CD4 +FoxP3 + regulatory T-cell manipulation in vivo: applications for allogeneic hematopoietic stem cell transplantation. Haematologica 2019; 104:1309-1321. [PMID: 31221786 PMCID: PMC6601084 DOI: 10.3324/haematol.2018.198838] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
CD4+FoxP3+ regulatory T cells (Tregs) are a non-redundant population critical for the maintenance of self-tolerance. Over the past decade, the use of these cells for therapeutic purposes in transplantation and autoimmune disease has emerged based on their capacity to inhibit immune activation. Basic science discoveries have led to identifying key receptors on Tregs that can regulate their proliferation and function. Notably, the understanding that IL-2 signaling is crucial for Treg homeostasis promoted the hypothesis that in vivo IL-2 treatment could provide a strategy to control the compartment. The use of low-dose IL-2 in vivo was shown to selectively expand Tregs versus other immune cells. Interestingly, a number of other Treg cell surface proteins, including CD28, CD45, IL-33R and TNFRSF members, have been identified which can also induce activation and proliferation of this population. Pre-clinical studies have exploited these observations to prevent and treat mice developing autoimmune diseases and graft-versus-host disease post-allogeneic hematopoietic stem cell transplantation. These findings support the development of translational strategies to expand Tregs in patients. Excitingly, the use of low-dose IL-2 for patients suffering from graft-versus-host disease and autoimmune disease has demonstrated increased Treg levels together with beneficial outcomes. To date, promising pre-clinical and clinical studies have directly targeted Tregs and clearly established the ability to increase their levels and augment their function in vivo. Here we review the evolving field of in vivo Treg manipulation and its application to allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
| | | | - Krishna V Komanduri
- Department of Microbiology and Immunology.,Sylvester Comprehensive Cancer Center.,Division of Transplantation and Cellular Therapy, Department of Medicine
| | - Robert B Levy
- Department of Microbiology and Immunology .,Division of Transplantation and Cellular Therapy, Department of Medicine.,Department of Ophthalmology, Miller School of Medicine, University of Miami, FL, USA
| |
Collapse
|
11
|
Geraghty NJ, Watson D, Sluyter R. Pharmacological blockade of the CD39/CD73 pathway but not adenosine receptors augments disease in a humanized mouse model of graft-versus-host disease. Immunol Cell Biol 2019; 97:597-610. [PMID: 30957314 DOI: 10.1111/imcb.12251] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 02/28/2019] [Accepted: 04/04/2019] [Indexed: 12/30/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation is a curative therapy for a number of hematological malignancies, but is limited by the development of graft-versus-host disease (GVHD). CD39 and CD73 form an ectoenzymatic pathway that hydrolyzes extracellular adenosine 5'-triphosphate (ATP) to adenosine, which respectively exacerbate or alleviate disease in allogeneic mouse models of GVHD. The current study aimed to explore the role of the CD39/CD73 pathway and adenosine receptor (AR) blockade in a humanized mouse model of GVHD. Immunodeficient nonobese diabetic-severe combined immunodeficiency-IL-2 receptor γnull mice were injected with human peripheral blood mononuclear cells, and subsequently injected with the CD39/CD73 antagonist αβ-methylene-ADP (APCP) (50 mg kg-1 ) or saline for 7 days, or the AR antagonist caffeine (10 mg kg-1 ) or saline for 14 days. Mice predominantly engrafted human CD4+ and CD8+ T cells, with smaller proportions of human regulatory T cells, invariant natural killer T cells, monocytes and dendritic cells. Neither APCP nor caffeine altered engraftment of these human leukocyte subsets. APCP (CD39/CD73 blockade) augmented GVHD as shown through increased weight loss and worsened liver histology, including increased leukocyte and human T-cell infiltration, and increased apoptosis. This treatment also increased serum human IL-2 concentrations and decreased the frequency of human CD39- CD73- CD4+ T cells. In contrast, caffeine (AR blockade) did not alter GVHD severity or human serum cytokine concentrations (IL-2, IL-6, IL-10 or tumor necrosis factor-α). In conclusion, blockade of CD39/CD73 but not ARs augments disease in a humanized mouse model of GVHD. These results indicate that CD39/CD73 blockade maintains sufficient extracellular ATP concentrations to promote GVHD in this model.
Collapse
Affiliation(s)
- Nicholas J Geraghty
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2252, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW, 2252, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW, 2252, Australia
| | - Debbie Watson
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2252, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW, 2252, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW, 2252, Australia
| | - Ronald Sluyter
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2252, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW, 2252, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW, 2252, Australia
| |
Collapse
|
12
|
Borcoman E, De La Rochere P, Richer W, Vacher S, Chemlali W, Krucker C, Sirab N, Radvanyi F, Allory Y, Pignot G, Barry de Longchamps N, Damotte D, Meseure D, Sedlik C, Bieche I, Piaggio E. Inhibition of PI3K pathway increases immune infiltrate in muscle-invasive bladder cancer. Oncoimmunology 2019; 8:e1581556. [PMID: 31069145 PMCID: PMC6492984 DOI: 10.1080/2162402x.2019.1581556] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/18/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Although immune checkpoint inhibitors have shown improvement in survival in comparison to chemotherapy in urothelial bladder cancer, many patients still fail to respond to these treatments and actual efforts are made to identify predictive factors of response to immunotherapy. Understanding the tumor-intrinsic molecular basis, like oncogenic pathways conditioning the presence or absence of tumor-infiltrating T cells (TILs), should provide a new rationale for improved anti-tumor immune therapies. In this study, we found that urothelial bladder cancer from human samples bearing PIK3CA gene mutations was significantly associated with lower expression of a defined immune gene signature, compared to unmutated ones. We identified a reduced 10-gene immune gene signature that discriminates muscle-invasive bladder cancer (MIBC) samples according to immune infiltration and PIK3CA mutation. Using a humanized mouse model, we observed that BKM120, a pan-PI3K inhibitor, significantly inhibited the growth of a human bladder cancer cell line bearing a PIK3CA mutation, associated to increased immune cell infiltration (hCD45+). Using qRT-PCR, we also found an increase in the expression of chemokines and immune genes in PIK3CA-mutated tumors from mice treated with BKM120, reflecting an active immune infiltrate in comparison to untreated ones. Moreover, the addition of BKM120 rendered PIK3CA-mutated tumors sensitive to PD-1 blockade. Our results provide a relevant rationale for combination strategies of PI3K inhibitors with immune checkpoint inhibitors to overcome resistance to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Edith Borcoman
- Department of Medical Oncology, Institut Curie, Paris, France.,Institut Curie, PSL Research University, INSERM U932, Paris, France
| | | | - Wilfrid Richer
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Sophie Vacher
- Department of Genetics, Institut Curie, Paris, France
| | | | | | - Nanour Sirab
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Francois Radvanyi
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Yves Allory
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France.,Department of diagnostic and theranostic medicine, Institut Curie, Saint-Cloud, France
| | - Géraldine Pignot
- Department of Urology, Institut Paoli-Calmettes, Marseille, France
| | | | - Diane Damotte
- Department of Pathology, Hôpital Cochin, Paris, France
| | - Didier Meseure
- Department of diagnostic and theranostic medicine, Institut Curie, Paris, France
| | - Christine Sedlik
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Ivan Bieche
- Department of Genetics, Institut Curie, Paris, France
| | - Eliane Piaggio
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| |
Collapse
|
13
|
Humanized Mice for the Study of Immuno-Oncology. Trends Immunol 2018; 39:748-763. [DOI: 10.1016/j.it.2018.07.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 01/28/2023]
|
14
|
Smaldini PL, Trejo F, Cohen JL, Piaggio E, Docena GH. Systemic IL-2/anti-IL-2Ab complex combined with sublingual immunotherapy suppresses experimental food allergy in mice through induction of mucosal regulatory T cells. Allergy 2018; 73:885-895. [PMID: 29319881 DOI: 10.1111/all.13402] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Therapeutic tolerance restoration has been proven to modify food allergy in patients and animal models and although sublingual immunotherapy (SLIT) has showed promise, combined therapy may be necessary to achieve a strong and long-term tolerance. AIMS In this work, we combined SLIT with systemic administration of IL-2 associated with an anti-IL-2 monoclonal antibody (IL-2/anti-IL-2Ab complex or IL-2C) to reverse the IgE-mediated experimental allergy. MATERIALS AND METHODS Balb/c mice were sensitized with cholera toxin and milk proteins and orally challenged with allergen to elicit hypersensitivity reactions. Then, allergic mice were treated with a sublingual administration of very low amounts of milk proteins combined with intraperitoneal injection of low doses of IL-2C. The animals were next re-exposed to allergens and mucosal as well as systemic immunological parameters were assessed in vivo and in vitro. RESULTS The treatment reduced serum specific IgE, IL-5 secretion by spleen cells and increased IL-10 and TGF-β in the lamina propria of buccal and duodenal mucosa. We found an augmented frequency of IL-10-secreting CD4+ CD25+ Foxp3+ regulatory T cells (Treg) in the submaxilar lymph nodes and buccal lamina propria. Tregs were sorted, characterized and adoptively transferred to naïve mice, which were subsequently sensitized. No allergy was experienced in these mice and we encouragingly discovered a faster and more efficient tolerance induction with the combined therapy compared with SLIT. CONCLUSION The combination of two therapeutic strategies rendered Treg-mediated tolerance more efficient compared to individual treatments and reversed the established IgE-mediated food allergy. This approach highlights the ability of IL-2C to expand Tregs, and it may represent a promising disease-modifying therapy for managing food allergy.
Collapse
Affiliation(s)
- P. L. Smaldini
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP); CONICET y Universidad Nacional de La Plata; La Plata Argentina
| | - F. Trejo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP); CONICET y Universidad Nacional de La Plata; La Plata Argentina
| | - J. L. Cohen
- Université Paris-Est; UMR_S955; UPEC; Créteil France
- Inserm, U955; Equipe 21; Créteil France
- Hôpital Henri Mondor; UPEC; APHP; Inserm; CIC Biothérapie; Créteil France
| | - E. Piaggio
- Institut Curie; PSL Research University; INSERM U932; Translational Immunotherapy team; Paris France
- Institut Curie; Centre d'Investigation Clinique Biothérapie CICBT 1428; Paris France
| | - G. H. Docena
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP); CONICET y Universidad Nacional de La Plata; La Plata Argentina
| |
Collapse
|
15
|
Salomon BL, Leclerc M, Tosello J, Ronin E, Piaggio E, Cohen JL. Tumor Necrosis Factor α and Regulatory T Cells in Oncoimmunology. Front Immunol 2018; 9:444. [PMID: 29593717 PMCID: PMC5857565 DOI: 10.3389/fimmu.2018.00444] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/19/2018] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor α (TNF) is a potent pro-inflammatory cytokine that has deleterious effect in some autoimmune diseases, which led to the use of anti-TNF drugs in some of these diseases. However, some rare patients treated with these drugs paradoxically develop an aggravation of their disease or new onset autoimmunity, revealing an immunosuppressive facet of TNF. A possible mechanism of this observation is the direct and positive effect of TNF on regulatory T cells (Tregs) through its binding to the TNF receptor type 2 (TNFR2). Indeed, TNF is able to increase expansion, stability, and possibly function of Tregs via TNFR2. In this review, we discuss the role of TNF in graft-versus-host disease as an example of the ambivalence of this cytokine in the pathophysiology of an immunopathology, highlighting the therapeutic potential of triggering TNFR2 to boost Treg expansion. We also describe new targets in immunotherapy of cancer, emphasizing on the putative suppressive effect of TNF in antitumor immunity and of the interest of blocking TNFR2 to regulate the Treg compartment.
Collapse
Affiliation(s)
- Benoît L Salomon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Mathieu Leclerc
- Université Paris-Est and INSERM U955, Créteil, France.,Service d'Hématologie Clinique et de Thérapie Cellulaire, Assistance Publique Hôpitaux de Paris (APHP), Hôpital H. Mondor, Créteil, France
| | - Jimena Tosello
- Center of Cancer Immunotherapy and Centre d'Investigation Clinique Biothérapie 1428, Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Emilie Ronin
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Eliane Piaggio
- Center of Cancer Immunotherapy and Centre d'Investigation Clinique Biothérapie 1428, Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - José L Cohen
- Université Paris-Est and INSERM U955, Créteil, France.,Centre d'Investigation Clinique Biothérapie, Assistance Publique Hôpitaux de Paris (APHP), Hôpital H. Mondor, Créteil, France
| |
Collapse
|
16
|
Hamdani S, Thiolat A, Naserian S, Grondin C, Moutereau S, Hulin A, Calderaro J, Grimbert P, Cohen JL, Azoulay D, Pilon C. Delayed and short course of rapamycin prevents organ rejection after allogeneic liver transplantation in rats. World J Gastroenterol 2017; 23:6962-6972. [PMID: 29097869 PMCID: PMC5658314 DOI: 10.3748/wjg.v23.i38.6962] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/06/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To test whether a delayed and short course of rapamycin would induce immunosuppressive effects following allogeneic orthotopic liver transplantation (OLT) in rats.
METHODS Allogeneic OLTs were performed using Dark Agouti livers transplanted into Lewis recipients, and syngeneic OLTs were performed using the Lewis rat strain. Rapamycin (1 mg/kg per day) was administered by gavage from day 4 to day 11 post-transplantation. Lymphocyte cellular compartments were analyzed by flow cytometry in draining lymph nodes, non-draining lymph nodes and the spleen at days 11 and 42 in rapamycin-treated rats, untreated control rats and syngeneic grafted rats. Skin grafts from Dark agouti or from F344 RT were performed at day 30 on liver grafted rats treated with rapamycin.
RESULTS An 8-d course of rapamycin treatment initiated 4 d following transplantation resulted in the survival of grafted rats for more than 100 d. In contrast, untreated rats died of liver failure within 13 to 21 d. The analysis of the cellular compartment revealed an increase in two cellular subpopulations, specifically myeloid-derived suppressor cells (MDSCs) and CD8+CD45RClow T cells, without major modifications in the regulatory T cell (Treg) compartment in treated rats in the early stages after grafting. We evaluated the ability of treated rats to reject third-party allogeneic skin grafts to confirm their immune competence. In contrast, when skin was collected from rats syngeneic to the grafted liver, it was not rejected.
CONCLUSION Our results demonstrate that short and delayed rapamycin treatment allows for tolerance in allogeneic OLT. The results also allowed for the identification of the mechanisms of tolerance induced by rapamycin by identifying MDSCs and CD8+CD45RClow T cells as associated with the state of tolerance.
Collapse
Affiliation(s)
- Salim Hamdani
- Université Paris-Est, UMR_S955, UPEC, Inserm, U955, Equipe 21, F-94000 Créteil, France
| | - Allan Thiolat
- Université Paris-Est, UMR_S955, UPEC, Inserm, U955, Equipe 21, F-94000 Créteil, France
| | - Sina Naserian
- Université Paris-Est, UMR_S955, UPEC, Inserm, U955, Equipe 21, F-94000 Créteil, France
| | - Cynthia Grondin
- Université Paris-Est, UMR_S955, UPEC, Inserm, U955, Equipe 21, F-94000 Créteil, France
| | - Stéphane Moutereau
- AP-HP, Laboratoire de Biochimie, Groupe Hospitalier Henri-Mondor Albert-Chenevier, F-94000 Créteil, France
| | - Anne Hulin
- AP-HP, Laboratoire de Pharmacologie-Toxicologie Biologiques, Groupe Hospitalier Henri-Mondor Albert-Chenevier, F-94000 Créteil, France
| | - Julien Calderaro
- AP-HP, Anatomie et Cytologie Pathologique, Groupe Hospitalier Henri-Mondor Albert-Chenevier, F-94000 Créteil, France
| | - Philippe Grimbert
- Université Paris-Est, UMR_S955, UPEC, Inserm, U955, Equipe 21, APHP, Inserm, CIC Biothérapie, Groupe Hospitalier Henri-Mondor Albert-Chenevier, F-94000 Créteil, France
| | - José Laurent Cohen
- Université Paris-Est, UMR_S955, UPEC, Inserm, U955, Equipe 21, APHP, Inserm, CIC Biothérapie, Groupe Hospitalier Henri-Mondor Albert-Chenevier, F-94000 Créteil, France
| | - Daniel Azoulay
- APHP, Service de Chirurgie HPB et Transplantation Hépatique, Groupe Hospitalier Henri-Mondor Albert-Chenevier, F-94000 Créteil, France
- Department of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Henri Mondor Hospital, 94010 Créteil, France
| | - Caroline Pilon
- Université Paris-Est, UMR_S955, UPEC, Inserm, U955, Equipe 21, APHP, Inserm, CIC Biothérapie, Groupe Hospitalier Henri-Mondor Albert-Chenevier, F-94000 Créteil, France
| |
Collapse
|
17
|
Cohen JL, Wood KJ. TNFR2: The new Treg switch? Oncoimmunology 2017; 7:e1373236. [PMID: 29296530 PMCID: PMC5739577 DOI: 10.1080/2162402x.2017.1373236] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 12/25/2022] Open
Abstract
Three recent publications identified the TNF/TNR2 pathway as a new target to reduce graft-versus-host-disease through regulatory T cells activation or to potentially switch on a strong anti-leukemic effect through regulatory T cells blockade in allogeneic hematopoietic stem cell transplantation. This identified the TNF/TNR2 pathway as a swith and as a new target for immune checkpoint therapy to modulate the immune regulation in this clinical setting.
Collapse
Affiliation(s)
- José L Cohen
- Université Paris-Est, Unité mixte de recherche S955, Université Paris Est Créteil, Créteil, France; INSERM, U955, Equipe 21, Créteil, France; Assistance Publique Hôpitaux de Paris (APHP), Hôpital H. Mondor-A. Chenevier, Centre d'Investigation Clinique Biothérapie, Créteil, France
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
18
|
Boieri M, Shah P, Dressel R, Inngjerdingen M. The Role of Animal Models in the Study of Hematopoietic Stem Cell Transplantation and GvHD: A Historical Overview. Front Immunol 2016; 7:333. [PMID: 27625651 PMCID: PMC5003882 DOI: 10.3389/fimmu.2016.00333] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022] Open
Abstract
Bone marrow transplantation (BMT) is the only therapeutic option for many hematological malignancies, but its applicability is limited by life-threatening complications, such as graft-versus-host disease (GvHD). The last decades have seen great advances in the understanding of BMT and its related complications; in particular GvHD. Animal models are beneficial to study complex diseases, as they allow dissecting the contribution of single components in the development of the disease. Most of the current knowledge on the therapeutic mechanisms of BMT derives from studies in animal models. Parallel to BMT, the understanding of the pathophysiology of GvHD, as well as the development of new treatment regimens, has also been supported by studies in animal models. Pre-clinical experimentation is the basis for deep understanding and successful improvements of clinical applications. In this review, we retrace the history of BMT and GvHD by describing how the studies in animal models have paved the way to the many advances in the field. We also describe how animal models contributed to the understanding of GvHD pathophysiology and how they are fundamental for the discovery of new treatments.
Collapse
Affiliation(s)
- Margherita Boieri
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Marit Inngjerdingen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
19
|
Control of GVHD by regulatory T cells depends on TNF produced by T cells and TNFR2 expressed by regulatory T cells. Blood 2016; 128:1651-9. [PMID: 27506541 DOI: 10.1182/blood-2016-02-700849] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/27/2016] [Indexed: 12/13/2022] Open
Abstract
Therapeutic CD4(+)Foxp3(+) natural regulatory T cells (Tregs) can control experimental graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HCT) by suppressing conventional T cells (Tconvs). Treg-based therapies are currently tested in clinical trials with promising preliminary results in allo-HCT. Here, we hypothesized that as Tregs are capable of modulating Tconv response, it is likely that the inflammatory environment and particularly donor T cells are also capable of influencing Treg function. Indeed, previous findings in autoimmune diabetes revealed a feedback mechanism that renders Tconvs able to stimulate Tregs by a mechanism that was partially dependent on tumor necrosis factor (TNF). We tested this phenomenon during alloimmune response in our previously described model of GVHD protection using antigen specific Tregs. Using different experimental approaches, we observed that control of GVHD by Tregs was fully abolished by blocking TNF receptor type 2 (TNFR2) or by using TNF-deficient donor T cells or TNFR2-deficient Tregs. Thus, our results show that Tconvs exert a powerful modulatory activity on therapeutic Tregs and clearly demonstrate that the sole defect of TNF production by donor T cells was sufficient to completely abolish the Treg suppressive effect in GVHD. Importantly, our findings expand the understanding of one of the central components of Treg action, the inflammatory context, and support that targeting TNF/TNFR2 interaction represents an opportunity to efficiently modulate alloreactivity in allo-HCT to either exacerbate it for a powerful antileukemic effect or reduce it to control GVHD.
Collapse
|
20
|
In Vivo Expansion of Regulatory T Cells by Low-Dose Interleukin-2 Treatment Increases Allograft Survival in Corneal Transplantation. Transplantation 2016; 100:525-32. [PMID: 26881788 DOI: 10.1097/tp.0000000000001044] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Corneal allograft survival dramatically decreases in hosts with inflamed or vascularized recipient beds. We have previously shown that in rejected corneal allografts regulatory T cells (Treg) demonstrate diminished Foxp3 expression and immunoregulatory function. Treatment with low doses of IL-2 selectively expands Treg and has been proposed for the treatment of autoimmune diseases. In this study, we investigated the effect of low-dose IL-2 administration on Treg function and corneal allograft survival. METHODS Allogeneic corneal transplantation was performed on inflamed host beds. Low-dose systemic IL-2 was administered starting 3 days before grafting until 6 weeks after transplantation. Frequencies of Treg and their immunosuppressive function and antigen specificity were assessed using flow cytometry, in vitro proliferation assays, and adoptive transfer experiments. Frequencies of effector T cells (Teff) and graft infiltrating immune cells were measured at 2 weeks posttransplantation. Long-term allograft survival was evaluated for up to 9 weeks using Kaplan-Meier survival analysis. RESULTS Treatment with low-dose IL-2 significantly increased frequencies of CD4CD25Foxp3 Treg and their immunosuppressive function. It also suppressed alloimmune response as shown by the decreased CD4 IFNγ T cell frequencies and graft infiltration of CD45 and CD4 cells. Clinical evaluation of the grafts showed significant improvement in long-term corneal allograft survival in the IL-2 treated group compared with controls. CONCLUSIONS Our study is the first to report that treatment with low-dose IL-2 increases survival of corneal allografts. We propose that IL-2-mediated Treg expansion can be an effective tool to prevent alloimmunity and to improve long-term allograft survival in transplantation.
Collapse
|
21
|
Mengel J, Cardillo F, Pontes-de-Carvalho L. Chronic Chagas' Disease: Targeting the Interleukin-2 Axis and Regulatory T Cells in a Condition for Which There Is No Treatment. Front Microbiol 2016; 7:675. [PMID: 27242702 PMCID: PMC4866556 DOI: 10.3389/fmicb.2016.00675] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 04/25/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jose Mengel
- Laboratory of Clinical Immunology, Oswaldo Cruz Institute (FIOCRUZ)Rio de Janeiro, Brazil; Laboratory of Clinical Immunology, Faculty of Medicine of Petrópolis-FASERio de Janeiro, Brazil
| | | | | |
Collapse
|
22
|
Kim N, Jeon YW, Nam YS, Lim JY, Im KI, Lee ES, Cho SG. Therapeutic potential of low-dose IL-2 in a chronic GVHD patient by in vivo expansion of regulatory T cells. Cytokine 2016; 78:22-6. [DOI: 10.1016/j.cyto.2015.11.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/17/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022]
|
23
|
Reduced interleukin-2 responsiveness impairs the ability of Treg cells to compete for IL-2 in nonobese diabetic mice. Immunol Cell Biol 2016; 94:509-19. [PMID: 26763864 DOI: 10.1038/icb.2016.7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/06/2016] [Accepted: 01/10/2015] [Indexed: 12/11/2022]
Abstract
Enhancement of regulatory T cell (Treg cell) frequency and function is the goal of many therapeutic strategies aimed at treating type 1 diabetes (T1D). The interleukin-2 (IL-2) pathway, which has been strongly implicated in T1D susceptibility in both humans and mice, is a master regulator of Treg cell homeostasis and function. We investigated how IL-2 pathway defects impact Treg cells in T1D-susceptible nonobese diabetic (NOD) mice in comparison with protected C57BL/6 and NOD congenic mice. NOD Treg cells were reduced in frequency specifically in the lymph nodes and expressed lower levels of CD25 and CD39/CD73 immunosuppressive molecules. In the spleen and blood, Treg cell frequency was preserved through expansion of CD25(low), effector phenotype Treg cells. Reduced CD25 expression led to decreased IL-2 signaling in NOD Treg cells. In vivo, treatment with IL-2-anti-IL-2 antibody complexes led to effective upregulation of suppressive molecules on NOD Treg cells in the spleen and blood, but had reduced efficacy on lymph node Treg cells. In contrast, NOD CD8(+) and CD4(+) effector T cells were not impaired in their response to IL-2 therapy. We conclude that NOD Treg cells have an impaired responsiveness to IL-2 that reduces their ability to compete for a limited supply of IL-2.
Collapse
|
24
|
Pérol L, Piaggio E. New Molecular and Cellular Mechanisms of Tolerance: Tolerogenic Actions of IL-2. Methods Mol Biol 2016; 1371:11-28. [PMID: 26530792 DOI: 10.1007/978-1-4939-3139-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Interleukin-2 (IL-2) is an old molecule with brand new functions. Indeed, IL-2 has been first described as a T-cell growth factor but recent data pointed out that its main function in vivo is the maintenance of immune tolerance. Mechanistically, IL-2 is essential for the development and function of CD4(+) Foxp3(+) regulatory T cells (Treg cells) that are essential players in the control of immune responded to self, tumors, microbes and grafts. Treg cells are exquisitely sensitive to IL-2 due to their constitutive expression of the high affinity IL-2 receptor (IL-2R) and the new paradigm suggests that low-doses of IL-2 could selectively boost Treg cells in vivo. Consequently, a growing body of clinical research is aiming at using IL-2 at low doses as a tolerogenic drug to boost endogenous Treg cells in patients suffering from autoimmune or inflammatory conditions. In this manuscript, we briefly review IL-2/IL-2R biology and the role of IL-2 in the development, maintenance, and function of Treg cells; and also its effects on other immune cell populations such as CD4(+) T helper cells and CD8(+) memory T cells. Then, focusing on type 1 diabetes, we review the preclinical studies and clinical trials supporting the use of low-doses IL-2 as a tolerogenic immunotherapy. Finally, we discuss the limitations and future directions for IL-2 based immunotherapy.
Collapse
Affiliation(s)
- Louis Pérol
- INSERM U932, 26 rue d'Ulm, 75005, Paris, France.
- Institut Curie, Section Recherche, 26 rue d'Ulm, 75005, Paris, France.
| | - Eliane Piaggio
- INSERM U932, 26 rue d'Ulm, 75005, Paris, France
- Institut Curie, Section Recherche, 26 rue d'Ulm, 75005, Paris, France
| |
Collapse
|
25
|
Henden AS, Hill GR. Cytokines in Graft-versus-Host Disease. THE JOURNAL OF IMMUNOLOGY 2015; 194:4604-12. [PMID: 25934923 DOI: 10.4049/jimmunol.1500117] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Graft-versus-host disease (GVHD) is a complication of allogeneic bone marrow transplantation whereby transplanted naive and marrow-derived T cells damage recipient tissue through similar mechanisms to those that allow destruction of malignant cells, the therapeutic intent of bone marrow transplantation. The manifestations and severity of GVHD are highly variable and are influenced by the proportions of naive cells maturing along regulatory T cell, Th1, Th2, or Th17 phenotypes. This maturation is largely influenced by local cytokines, which, in turn, activate transcription factors and drive development toward a dominant phenotype. In addition, proinflammatory cytokines exert direct effects on GVHD target tissues. Our knowledge of the role that cytokines play in orchestrating GVHD is expanding rapidly and parallels other infective and inflammatory conditions in which a predominant T cell signature is causative of pathology. Because a broad spectrum of cytokine therapies is now routinely used in clinical practice, they are increasingly relevant to transplant medicine.
Collapse
Affiliation(s)
- Andrea S Henden
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4006, Queensland, Australia; and The Royal Brisbane and Women's Hospital, Brisbane 4029, Queensland, Australia
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4006, Queensland, Australia; and The Royal Brisbane and Women's Hospital, Brisbane 4029, Queensland, Australia
| |
Collapse
|