1
|
Ohm B, Giannou AD, Harriman D, Oh J, Jungraithmayr W, Zazara DE. Chimerism and immunological tolerance in solid organ transplantation. Semin Immunopathol 2025; 47:27. [PMID: 40387984 PMCID: PMC12089243 DOI: 10.1007/s00281-025-01052-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
In solid organ transplantation, chimerism inevitably occurs via the coexistence of donor-derived cells from the graft and host cells throughout the recipient. However, long-term immunosuppressive treatment is needed to suppress host immune responses to the foreign organ graft. The deliberate induction of stable mixed bone marrow chimerism to achieve donor-specific immunological tolerance in solid organ graft recipients is an ambitious goal that may significantly contribute to the long-term survival of solid organ grafts and their recipients. While this strategy has been effectively established in laboratory animals and some promising clinical case series have been reported, widespread clinical application is still limited by the toxicity of the necessary conditioning regimens. On the other hand, the naturally occurring chimeric state resulting from the bidirectional transplacental cell trafficking during pregnancy, the so-called feto-maternal microchimerism, can also induce immune tolerance and thus influence the outcome of mother-to-child or child-to-mother organ transplantation. This review provides an overview of the field's historical development, clinical results, and underlying principles of (micro) chimerism-based tolerance.
Collapse
Affiliation(s)
- Birte Ohm
- Department of Thoracic Surgery, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Anastasios D Giannou
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- General Surgery, Liver, Pancreas and Intestinal Transplant Unit, Hospital Universitario-Fundación Favaloro, Buenos Aires, Argentina
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - David Harriman
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Jun Oh
- University Children's Hospital, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Wolfgang Jungraithmayr
- Department of Thoracic Surgery, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
- Division of Thoracic Surgery, Rostock University Medical Center, Rostock, Germany
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Dimitra E Zazara
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- University Children's Hospital, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Division for Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
2
|
Linhares AFS, Rezende BM, Castor MGM. Allogeneic Mouse Models of Graft-Versus-Host Disease. Methods Mol Biol 2025; 2907:127-140. [PMID: 40100596 DOI: 10.1007/978-1-0716-4430-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Graft-versus-host disease (GVHD) is a serious and potentially fatal complication that occurs after allogeneic hematopoietic cell transplantation. It is characterized by the immune response of the graft (from the donor) against the tissue of host (recipient), resulting in damage to target organs such as the skin, liver, and intestines. Understanding the underlying mechanisms of GVHD is crucial for improving treatment and prevention strategies for this debilitating condition. Animal experimental models play a key role in this process, allowing for the investigation of immune mechanisms and the evaluation of new therapies. These models provide valuable insights into the pathophysiology of GVHD and help develop more effective therapeutic approaches to improve clinical outcomes in transplant patients. In this chapter, we provide a brief overview of allogeneic and semi-allogeneic models of GVHD in mice. Then, we will address the step-by-step process for inducing experimental GVHD in mice using an allogeneic model in mice with total body irradiation myeloablation.
Collapse
Affiliation(s)
- Ana Flávia Santos Linhares
- Graduate Program in Biological Sciences: Physiology and Pharmacology, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Barbara Maximino Rezende
- Department of Basic Nurse of Nurse's School, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marina Gomes Miranda Castor
- Graduate Program in Biological Sciences: Physiology and Pharmacology, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
3
|
Suek N, Young T, Fu J. Immune cell profiling in intestinal transplantation. Hum Immunol 2024; 85:110808. [PMID: 38762429 PMCID: PMC11283363 DOI: 10.1016/j.humimm.2024.110808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/20/2024]
Abstract
Since the first published case study of human intestinal transplantation in 1967, there have been significant studies of intestinal transplant immunology in both animal models and humans. An improved understanding of the profiles of different immune cell subsets is critical for understanding their contributions to graft outcomes. While different studies have focused on the contribution of one or a few subsets to intestinal transplant, no study has integrated these data for a comprehensive overview of immune dynamics after intestinal transplant. Here, we provide a systematic review of the literature on different immune subsets and discuss their roles in intestinal transplant outcomes on multiple levels, focusing on chimerism and graft immune reconstitution, clonal alloreactivity, and cell phenotype. In Sections 1, 2 and 3, we lay out a shared framework for understanding intestinal transplant, focusing on the mechanisms of rejection or tolerance in the context of mucosal immunology and illustrate the unique role of the bidirectional graft-versus-host (GvH) and host-versus-graft (HvG) alloresponse. In Sections 4, 5 and 6, we further expand upon these concepts as we discuss the contribution of different cell subsets to intestinal transplant. An improved understanding of intestinal transplantation immunology will bring us closer to maximizing the potential of this important treatment.
Collapse
Affiliation(s)
- Nathan Suek
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Tyla Young
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
4
|
Sykes M. Tolerance in intestinal transplantation. Hum Immunol 2024; 85:110793. [PMID: 38580539 PMCID: PMC11144570 DOI: 10.1016/j.humimm.2024.110793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/07/2024]
Abstract
Intestinal transplantation (ITx) is highly immunogenic, resulting in the need for high levels of immunosuppression, with frequent complications along with high rejection rates. Tolerance induction would provide a solution to these limitations. Detailed studies of alloreactive T cell clones as well as multiparameter flow cytometry in the graft and peripheral tissues have provided evidence for several tolerance mechanisms that occur spontaneously following ITx, which might provide targets for further interventions. These include the frequent occurrence of macrochimerism and engraftment in the recipient bone marrow of donor hematopoietic stem and progenitor cells carried in the allograft. These phenomena are seen most frequently in recipients of multivisceral transplants and are associated with reduced rejection rates. They reflect powerful graft-vs-host responses that enter the peripheral lymphoid system and bone marrow after expanding within and emigrating from the allograft. Several mechanisms of tolerance that may result from this lymphohematopoietic graft-vs-host response are discussed. Transcriptional profiling in quiescent allografts reveals tolerization of pre-existing host-vs-graft-reactive T cells that enter the allograft mucosa and become tissue-resident memory cells. Dissection of the pathways driving and maintaining this tolerant tissue-resident state among donor-reactive T cells will allow controlled tolerance induction through specific therapeutic approaches.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Department of Microbiology and Immunology and Department of Surgery, Columbia University, New York, NY, USA.
| |
Collapse
|
5
|
Xiao Y, Schroeter A, Martin F, Matsunaga T, Nakamori K, Roesel MJ, Habal M, Chong AS, Zhou H, Tullius SG. Sex as a biological variable: Mechanistic insights and clinical relevance in solid organ transplantation. Am J Transplant 2023; 23:1661-1672. [PMID: 37543092 PMCID: PMC10838351 DOI: 10.1016/j.ajt.2023.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 08/07/2023]
Abstract
Biological sex affects immunity broadly, with recognized effects on the incidence and severity of autoimmune diseases, infections, and malignancies. Consequences of sex on alloimmunity and outcomes in solid organ transplantation are less well defined. Clinical studies have shown that donor and recipient sex independently impact transplant outcomes, which are further modified by aging. Potential mechanisms have thus far not been detailed and may include hormonal, genetic, and epigenetic components. Here, we summarize relevant findings in immunity in addition to studies in clinical and experimental organ transplantation detailing the effects of biological sex on alloimmunity. Understanding both clinical impact and mechanisms is expected to provide critical insights on the complexity of alloimmune responses, with the potential to fine-tune treatment and allocation while providing a rationale to include both sexes in transplant research.
Collapse
Affiliation(s)
- Yao Xiao
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andreas Schroeter
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Regenerative Medicine and Experimental Surgery, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Friederike Martin
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Surgery, CVK/CCM, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Tomohisa Matsunaga
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Keita Nakamori
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Maximilian J Roesel
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Marlena Habal
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, Columbia University, New York, New York, USA
| | - Anita S Chong
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Hao Zhou
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
6
|
Sykes M. Leveraging the lymphohematopoietic graft-versus-host reaction (LGVHR) to achieve allograft tolerance and restore self tolerance with minimal toxicity. IMMUNOTHERAPY ADVANCES 2023; 3:ltad008. [PMID: 37426630 PMCID: PMC10327628 DOI: 10.1093/immadv/ltad008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/12/2023] [Indexed: 07/11/2023] Open
Abstract
Mixed allogeneic chimerism has considerable potential to advance the achievement of immune tolerance to alloantigens for transplantation and the restoration of self-tolerance in patients with autoimmune disease. In this article, I review evidence that graft-versus-host (GVH) alloreactivity without graft-vs-host disease (GVHD), termed a lymphohematopoietic graft-vs-host reaction (LGVHR), can promote the induction of mixed chimerism with minimal toxicity. LGVHR was originally shown to occur in an animal model when non-tolerant donor lymphocytes were administered to mixed chimeras in the absence of inflammatory stimuli and was found to mediate powerful graft-vs-leukemia/lymphoma effects without GVHD. Recent large animal studies suggest a role for LGVHR in promoting durable mixed chimerism and the demonstration that LGVHR promotes chimerism in human intestinal allograft recipients has led to a pilot study aiming to achieve durable mixed chimerism.
Collapse
Affiliation(s)
- Megan Sykes
- Correspondence: Megan Sykes, Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, 650 West 168th Street, Suite 1512, New York, NY 10032, USA.
| |
Collapse
|
7
|
Ash S, Askenasy N. Immunotherapy for neuroblastoma by hematopoietic cell transplantation and post-transplant immunomodulation. Crit Rev Oncol Hematol 2023; 185:103956. [PMID: 36893946 DOI: 10.1016/j.critrevonc.2023.103956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/14/2022] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroblastoma represents a relatively common childhood tumor that imposes therapeutic difficulties. High risk neuroblastoma patients have poor prognosis, display limited response to radiochemotherapy and may be treated by hematopoietic cell transplantation. Allogeneic and haploidentical transplants have the distinct advantage of reinstitution of immune surveillance, reinforced by antigenic barriers. The key factors favorable to ignition of potent anti-tumor reactions are transition to adaptive immunity, recovery from lymphopenia and removal of inhibitory signals that inactivate immune cells at the local and systemic levels. Post-transplant immunomodulation may further foster anti-tumor reactivity, with positive but transient impact of infusions of lymphocytes and natural killer cells both from the donor, the recipient or third party. The most promising approaches include introduction of antigen-presenting cells in early post-transplant stages and neutralization of inhibitory signals. Further studies will likely shed light on the nature and actions of suppressor factors within tumor stroma and at the systemic level.
Collapse
Affiliation(s)
- Shifra Ash
- Department of Pediatric Hematology-Oncology, Rambam Medical Center, Haifa, Israel; Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Nadir Askenasy
- Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
8
|
Anti- E. coli Immunoglobulin Yolk (IgY): Reduction of pathogen receptors and inflammation factors could be caused by decrease in E. coli load. Heliyon 2023; 9:e13876. [PMID: 36873547 PMCID: PMC9982617 DOI: 10.1016/j.heliyon.2023.e13876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Graft versus host disease (GVHD) remains the major cause of morbidity and mortality after allogeneic stem cell transplantation, especially for intestinal GVHD, as steroid resistant GVHD results in high mortality. For this reason, new treatments of GVHD are needed. One approach is the reduction of pathogenic bacteria using anti-E. coli Immunoglobulin Yolk (IgY). In a haploidentical murine model, B6D2F1 mice conditioned with total body irradiation (TBI), received bone marrow cells (BM) and splenocytes (SC) from either syngeneic (Syn = B6D2F1) or allogeneic (Allo = C57BL/6) donors. Following this, animals received from day -2 until day +28 chow contained IgY or control chow. Thereafter the incidence and severity of aGVHD, the cytokines, chemokines, IDO1 and different pathogen-recognition receptors (PRR) were analyzed and compared to control animals (received chow without IgY). We found that animals receiving chow with IgY antibody showed reduced GVHD severity compared to control animals. On day28 after alloBMT, IDO, NOD2, TLR2, TLR4 and the inflammatory chemokine CCL3, were reduced in the colon and correlated with a significant decrease in E. coli bacteria. In summary chow containing chicken antibodies (IgY) improved GVHD via decrease in bacterial load of E coli conducting to reduction of pathogen receptors (NOD2, TLR2 and 4), IDO, chemokines and cytokines.
Collapse
|
9
|
Socie G, Michonneau D. Milestones in acute GVHD pathophysiology. Front Immunol 2022; 13:1079708. [PMID: 36544776 PMCID: PMC9760667 DOI: 10.3389/fimmu.2022.1079708] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/22/2022] [Indexed: 12/07/2022] Open
Abstract
In the past 65 years, over 25 000 referenced articles have been published on graft-versus-host disease (GVHD). Although this included clinically orientated papers or publications on chronic GVHD, the conservative estimate of scientific publications still contains several thousands of documents on the pathophysiology of acute GVHD. Thus, summarizing what we believe are prominent publications that can be considered milestones in our knowledge of this disease is a challenging and inherently biased task. Here we review from a historical perspective what can be regarded as publications that have made the field move forward. We also included several references of reviews on aspects we could not cover in detail.
Collapse
Affiliation(s)
- Gerard Socie
- Université Paris Cité, Paris, France
- APHP, Hématologie Greffe, Hôpital Saint Louis, Paris, France
- INSERM UMR 976, Hôpital Saint Louis, Paris, France
| | - David Michonneau
- Université Paris Cité, Paris, France
- APHP, Hématologie Greffe, Hôpital Saint Louis, Paris, France
- INSERM UMR 976, Hôpital Saint Louis, Paris, France
| |
Collapse
|
10
|
Song Q, Nasri U, Nakamura R, Martin PJ, Zeng D. Retention of Donor T Cells in Lymphohematopoietic Tissue and Augmentation of Tissue PD-L1 Protection for Prevention of GVHD While Preserving GVL Activity. Front Immunol 2022; 13:907673. [PMID: 35677056 PMCID: PMC9168269 DOI: 10.3389/fimmu.2022.907673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (Allo-HCT) is a curative therapy for hematological malignancies (i.e., leukemia and lymphoma) due to the graft-versus-leukemia (GVL) activity mediated by alloreactive T cells that can eliminate residual malignant cells and prevent relapse. However, the same alloreactive T cells can cause a serious side effect, known as graft-versus-host disease (GVHD). GVHD and GVL occur in distinct organ and tissues, with GVHD occurring in target organs (e.g., the gut, liver, lung, skin, etc.) and GVL in lympho-hematopoietic tissues where hematological cancer cells primarily reside. Currently used immunosuppressive drugs for the treatment of GVHD inhibit donor T cell activation and expansion, resulting in a decrease in both GVHD and GVL activity that is associated with cancer relapse. To prevent GVHD, it is important to allow full activation and expansion of alloreactive T cells in the lympho-hematopoietic tissues, as well as prevent donor T cells from migrating into the GVHD target tissues, and tolerize infiltrating T cells via protective mechanisms, such as PD-L1 interacting with PD-1, in the target tissues. In this review, we will summarize major approaches that prevent donor T cell migration into GVHD target tissues and approaches that augment tolerization of the infiltrating T cells in the GVHD target tissues while preserving strong GVL activity in the lympho-hematopoietic tissues.
Collapse
Affiliation(s)
- Qingxiao Song
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Fujian Medical University Center of Translational Hematology, Fujian Institute of Hematology, and Fujian Medical University Union Hospital, Fuzhou, China
| | - Ubaydah Nasri
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| | - Ryotaro Nakamura
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| | - Paul J Martin
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, United States
| | - Defu Zeng
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| |
Collapse
|
11
|
Scheurer J, Leithäuser F, Debatin KM, Strauss G. Modeling acute graft-versus-host disease (aGVHD) in murine bone marrow transplantation (BMT) models with MHC disparity. Methods Cell Biol 2022; 168:19-39. [PMID: 35366982 DOI: 10.1016/bs.mcb.2021.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
For more than 50years, hematopoietic stem cell transplantation (HSCT) has been the major curative therapy for hematological malignancies and genetic disorders, but its success is limited by the development of graft-versus-host disease (GVHD). GVHD represents a post-transplantation disorder representing the immune-mediated attack of transplant-derived T cells against recipient tissue finally leading to increased morbidity and mortality of the recipient. GVHD develops if donor and recipient are disparate in major or minor histocompatibility antigens (MHC, miHA). Most of the initial knowledge about the biology of GVHD is derived from murine bone marrow transplantation (BMT) models. Of course, GVHD mouse models do not reflect one to one the human situation, but they contribute significantly to our understanding how conditioning and danger signals activate the immune system, enlighten the role of individual molecules, e.g., cytokines, chemokines, death-inducing ligands, define the function of lymphocytes subpopulations for GVHD development and have significant impact on establishing new treatment and prevention strategies used in clinical HSCT. This chapter describes in detail the procedure of allogeneic BMT and the development of GVHD in two commonly used allogeneic murine BMT models (B6→B6.bm1, B6→B6D2F1) with different MHC disparities, which can be used as a basis for advanced studies of GVHD pathology or the development of new treatment strategies.
Collapse
Affiliation(s)
- Jasmin Scheurer
- University Medical Center Ulm, Department of Pediatrics and Adolescent Medicine, Ulm, Germany
| | | | - Klaus-Michael Debatin
- University Medical Center Ulm, Department of Pediatrics and Adolescent Medicine, Ulm, Germany
| | - Gudrun Strauss
- University Medical Center Ulm, Department of Pediatrics and Adolescent Medicine, Ulm, Germany.
| |
Collapse
|
12
|
Fu J, Zuber J, Shonts B, Obradovic A, Wang Z, Frangaj K, Meng W, Rosenfeld AM, Waffarn EE, Liou P, Lau SP, Savage TM, Yang S, Rogers K, Danzl NM, Ravella S, Satwani P, Iuga A, Ho SH, Griesemer A, Shen Y, Prak ETL, Martinez M, Kato T, Sykes M. Lymphohematopoietic graft-versus-host responses promote mixed chimerism in patients receiving intestinal transplantation. J Clin Invest 2021; 131:141698. [PMID: 33630757 DOI: 10.1172/jci141698] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 02/23/2021] [Indexed: 12/22/2022] Open
Abstract
In humans receiving intestinal transplantation (ITx), long-term multilineage blood chimerism often develops. Donor T cell macrochimerism (≥4%) frequently occurs without graft-versus-host disease (GVHD) and is associated with reduced rejection. Here we demonstrate that patients with macrochimerism had high graft-versus-host (GvH) to host-versus-graft (HvG) T cell clonal ratios in their allografts. These GvH clones entered the circulation, where their peak levels were associated with declines in HvG clones early after transplant, suggesting that GvH reactions may contribute to chimerism and control HvG responses without causing GVHD. Consistently, donor-derived T cells, including GvH clones, and CD34+ hematopoietic stem and progenitor cells (HSPCs) were simultaneously detected in the recipients' BM more than 100 days after transplant. Individual GvH clones appeared in ileal mucosa or PBMCs before detection in recipient BM, consistent with an intestinal mucosal origin, where donor GvH-reactive T cells expanded early upon entry of recipient APCs into the graft. These results, combined with cytotoxic single-cell transcriptional profiles of donor T cells in recipient BM, suggest that tissue-resident GvH-reactive donor T cells migrated into the recipient circulation and BM, where they destroyed recipient hematopoietic cells through cytolytic effector functions and promoted engraftment of graft-derived HSPCs that maintain chimerism. These mechanisms suggest an approach to achieving intestinal allograft tolerance.
Collapse
Affiliation(s)
- Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Julien Zuber
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Brittany Shonts
- Columbia Center for Translational Immunology, Department of Medicine and
| | | | - Zicheng Wang
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, New York, USA
| | - Kristjana Frangaj
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Wenzhao Meng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Aaron M Rosenfeld
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Sai-Ping Lau
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Thomas M Savage
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Suxiao Yang
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Kortney Rogers
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Nichole M Danzl
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Shilpa Ravella
- Division of Digestive and Liver Diseases, Department of Medicine
| | | | - Alina Iuga
- Department of Pathology and Cell Biology, and
| | - Siu-Hong Ho
- Columbia Center for Translational Immunology, Department of Medicine and
| | - Adam Griesemer
- Columbia Center for Translational Immunology, Department of Medicine and.,Department of Surgery
| | - Yufeng Shen
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, New York, USA
| | - Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine and.,Department of Surgery.,Department of Microbiology and Immunology, Columbia University, New York, New York, USA
| |
Collapse
|
13
|
Bäuerlein CA, Qureischi M, Mokhtari Z, Tabares P, Brede C, Jordán Garrote AL, Riedel SS, Chopra M, Reu S, Mottok A, Arellano-Viera E, Graf C, Kurzwart M, Schmiedgen K, Einsele H, Wölfl M, Schlegel PG, Beilhack A. A T-Cell Surface Marker Panel Predicts Murine Acute Graft-Versus-Host Disease. Front Immunol 2021; 11:593321. [PMID: 33584657 PMCID: PMC7880247 DOI: 10.3389/fimmu.2020.593321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/11/2020] [Indexed: 11/13/2022] Open
Abstract
Acute graft-versus-host disease (aGvHD) is a severe and often life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT). AGvHD is mediated by alloreactive donor T-cells targeting predominantly the gastrointestinal tract, liver, and skin. Recent work in mice and patients undergoing allo-HCT showed that alloreactive T-cells can be identified by the expression of α4β7 integrin on T-cells even before manifestation of an aGvHD. Here, we investigated whether the detection of a combination of the expression of T-cell surface markers on peripheral blood (PB) CD8+ T-cells would improve the ability to predict aGvHD. To this end, we employed two independent preclinical models of minor histocompatibility antigen mismatched allo-HCT following myeloablative conditioning. Expression profiles of integrins, selectins, chemokine receptors, and activation markers of PB donor T-cells were measured with multiparameter flow cytometry at multiple time points before the onset of clinical aGvHD symptoms. In both allo-HCT models, we demonstrated a significant upregulation of α4β7 integrin, CD162E, CD162P, and conversely, a downregulation of CD62L on donor T-cells, which could be correlated with the development of aGvHD. Other surface markers, such as CD25, CD69, and CC-chemokine receptors were not found to be predictive markers. Based on these preclinical data from mouse models, we propose a surface marker panel on peripheral blood T-cells after allo-HCT combining α4β7 integrin with CD62L, CD162E, and CD162P (cutaneous lymphocyte antigens, CLA, in humans) to identify patients at risk for developing aGvHD early after allo-HCT.
Collapse
Affiliation(s)
- Carina A Bäuerlein
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Musga Qureischi
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Zeinab Mokhtari
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany
| | - Paula Tabares
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany
| | - Christian Brede
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Ana-Laura Jordán Garrote
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Simone S Riedel
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Martin Chopra
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany
| | - Simone Reu
- Institute of Pathology, Würzburg University, Würzburg, Germany
| | - Anja Mottok
- Institute of Pathology, Würzburg University, Würzburg, Germany
| | - Estibaliz Arellano-Viera
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany
| | - Carolin Graf
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany
| | - Miriam Kurzwart
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany
| | - Katharina Schmiedgen
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany
| | - Hermann Einsele
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Matthias Wölfl
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany.,Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Paul-Gerhardt Schlegel
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany.,Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Würzburg University, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| |
Collapse
|
14
|
|
15
|
Zhou Y, Cao L, Guo H, Hong Y, Wang M, Wang K, Huang X, Chang Y. Th2 polarization in target organs is involved in the alleviation of pathological damage mediated by transplanting granulocyte colony-stimulating factor-primed donor T cells. SCIENCE CHINA-LIFE SCIENCES 2020; 64:1087-1096. [PMID: 32880861 DOI: 10.1007/s11427-020-1754-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/09/2020] [Indexed: 11/24/2022]
Abstract
Acute graft-versus-host disease (aGVHD) is caused by allo-activated donor T cells infiltrating target organs. As a regulator of immune function, granulocyte colony-stimulating factor (G-CSF) has been demonstrated to relieve the aGVHD reaction. However, the role of G-CSF-primed donor T cells in specific target organs is still unknown. In this study, we employed a classical MHC-mismatched transplantation mouse model (C57BL/6 into BALB/c) and found that recipient mice transplanted with G-CSF-primed T cells exhibited prolonged survival compared with that of the PBS-treated group. This protective function against GVHD mediated by G-CSF-primed donor T cells was further confirmed by decreased clinical and pathological scores in this aGVHD mouse model, especially in the lung and gut. Moreover, we found that T cells polarized towards Th2 cells and regulatory T cells were increased in specific target organs. In addition, G-CSF treatment inhibited inducible co-stimulator (ICOS) expression and increased the expression of tolerance-related genes in recipient mice. Our study provides new insight into the immune regulatory effects of G-CSF on T cell-mediated aGVHD, especially for its precise regulation in GVHD target organs.
Collapse
Affiliation(s)
- Yang Zhou
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, Peking University, Beijing, 100044, China
| | - Leqing Cao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, Peking University, Beijing, 100044, China
| | - Huidong Guo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, Peking University, Beijing, 100044, China
| | - Yan Hong
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, Peking University, Beijing, 100044, China
| | - Ming Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, Peking University, Beijing, 100044, China
| | - Ke Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, Peking University, Beijing, 100044, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, Peking University, Beijing, 100044, China. .,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100044, China.
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, Peking University, Beijing, 100044, China.
| |
Collapse
|
16
|
Alvarez-Laderas I, Ramos TL, Medrano M, Caracuel-García R, Barbado MV, Sánchez-Hidalgo M, Zamora R, Alarcón-de-la-Lastra C, Hidalgo FJ, Piruat JI, Caballero-Velázquez T, Pérez-Simón JA. Polyphenolic Extract (PE) from Olive Oil Exerts a Potent Immunomodulatory Effect and Prevents Graft-versus-Host Disease in a Mouse Model. Biol Blood Marrow Transplant 2019; 26:615-624. [PMID: 31756538 DOI: 10.1016/j.bbmt.2019.11.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 09/22/2019] [Accepted: 11/14/2019] [Indexed: 12/25/2022]
Abstract
Polyphenols are a group of chemical substances found in plants, with immunomodulatory, antiproliferative, and anti-inflammatory properties that might be useful in the prophylaxis and treatment of graft-versus-host disease (GVHD). Polyphenolic extract (PE) obtained from extra virgin olive oil (EVOO) decreased the activation and proliferation of activated T cells. In addition, a decreased production of proinflammatory cytokines was observed upon exposure to PE. Western blot assays showed a marked inhibition of Akt phosphorylation and nuclear translocation of NF-κB in activated T cells. In a murine model of acute GVHD, we observed that mice that received a diet supplemented in PE (600 ppm) presented a higher survival rate and lower risk of developing GVHD when compared with the group that received a control diet. Histopathologic examination showed a significantly lower gut involvement in mice receiving PE, with a decrease in proinflammatory cytokines (IL-2, IL-17, and TNF-α) in serum and the reestablishment of butyrate concentration in the gut. In conclusion, PE obtained from EVOO exerted a potent immunomodulatory effect, reducing the activation and proliferation of activated T cells and the production of proinflammatory cytokines. In a murine model of acute GVHD, a PE-supplemented diet reduced the incidence and severity of the disease and increased survival after transplantation.
Collapse
Affiliation(s)
- Isabel Alvarez-Laderas
- Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - Teresa L Ramos
- Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - Mayte Medrano
- Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - Rocío Caracuel-García
- Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - María Victoria Barbado
- Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | | | - Rosario Zamora
- Instituto de la Grasa, Consejo Superior de Investigaciones Científicas, Campus Universitario-Edificio 46, Seville, Spain
| | | | - Francisco J Hidalgo
- Instituto de la Grasa, Consejo Superior de Investigaciones Científicas, Campus Universitario-Edificio 46, Seville, Spain
| | - José Ignacio Piruat
- Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - Teresa Caballero-Velázquez
- Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - José Antonio Pérez-Simón
- Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain.
| |
Collapse
|
17
|
Wajant H, Beilhack A. Targeting Regulatory T Cells by Addressing Tumor Necrosis Factor and Its Receptors in Allogeneic Hematopoietic Cell Transplantation and Cancer. Front Immunol 2019; 10:2040. [PMID: 31555271 PMCID: PMC6724557 DOI: 10.3389/fimmu.2019.02040] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/12/2019] [Indexed: 12/15/2022] Open
Abstract
An intricate network of molecular and cellular actors orchestrates the delicate balance between effector immune responses and immune tolerance. The pleiotropic cytokine tumor necrosis factor-alpha (TNF) proves as a pivotal protagonist promoting but also suppressing immune responses. These opposite actions are accomplished through specialist cell types responding to TNF via TNF receptors TNFR1 and TNFR2. Recent findings highlight the importance of TNFR2 as a key regulator of activated natural FoxP3+ regulatory T cells (Tregs) in inflammatory conditions, such as acute graft-vs.-host disease (GvHD) and the tumor microenvironment. Here we review recent advances in our understanding of TNFR2 signaling in T cells and discuss how these can reconcile seemingly conflicting observations when manipulating TNF and TNFRs. As TNFR2 emerges as a new and attractive target we furthermore pinpoint strategies and potential pitfalls for therapeutic targeting of TNFR2 for cancer treatment and immune tolerance after allogeneic hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany.,Else-Kröner-Forschungskolleg Würzburg, Würzburg University Hospital, Würzburg University, Würzburg, Germany
| |
Collapse
|
18
|
Abstract
This review focuses on our recent studies involving nonmyeloablative bone marrow transplantation as an approach to inducing organ allograft tolerance across MHC barriers in nonhuman primates and in patients. The clinical studies are focused on mechanisms of tolerance involved in a protocol carried out at Massachusetts General Hospital in HLA-mismatched haploidentical combinations for the induction of renal allograft tolerance. These studies, in which chimerism was only transient and GVHD did not occur, suggest an early role for donor-specific regulatory T cells in tolerance induction, followed by partial and gradual deletion of donor-reactive T cells. We utilized high-throughput sequencing methodologies in a novel way to identify and track large numbers of alloreactive T cell receptors (TCRs). This method has been shown to identify biologically significant alloreactive TCRs in transplant patients and pointed to clonal deletion as a major mechanism of long-term tolerance in these patients. More recently, we adapted this sequencing method to optimally identify the donor-specific regulatory T cell (Treg) repertoire. Interrogation of the early posttransplant repertoire demonstrated expansion of donor-specific Tregs in association with tolerance. Our studies suggest a role for the kidney graft in tolerance by these mechanisms in patients who had only transient chimerism. Nonhuman primate studies indicate that other organs, including the heart, the lungs and the liver, are less readily tolerated following a period of transient mixed chimerism. Our efforts to extend the reach of mixed chimerism for tolerance induction beyond the kidney are therefore focused on the addition of recipient Tregs to the protocol. This approach has the potential to enhance chimerism while further reducing the risk of GVHD.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Medical Center, New York, NY, USA.
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Medical Center, New York, NY, USA.
| | - Adam D Griesemer
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
19
|
Cassady K, Martin PJ, Zeng D. Regulation of GVHD and GVL Activity via PD-L1 Interaction With PD-1 and CD80. Front Immunol 2018; 9:3061. [PMID: 30622541 PMCID: PMC6308317 DOI: 10.3389/fimmu.2018.03061] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for hematological malignancies (i.e. leukemia and lymphoma), because graft-versus-leukemia (GVL) activity mediated by alloreactive T cells can eliminate residual malignant cells and prevent relapse. However, the same alloreactive T cells also mediate a severe side effect, graft-versus-host disease (GVHD), and prevention of GVHD while preserving GVL activity remains an elusive goal. The immune checkpoint molecule PD-L1 and its interaction with PD-1 receptor in regulating cancer immunity is under intensive and wide-spread study, but knowledge about this interaction in regulating GVHD and GVL activity is very limited. In this review, we summarize the literature exploring how PD-L1 interaction with its receptors PD-1 and CD80 regulate GVHD and GVL activities, how PD-L1 signaling regulates T cell metabolic profiles, and how a differential role of PD-L1 interaction with PD-1, CD80 or both may provide a novel avenue to prevent GVHD while preserving strong GVL effects.
Collapse
Affiliation(s)
- Kaniel Cassady
- Irell and Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, United States.,Department of Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, CA, United States.,Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Paul J Martin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Medicine, University of Washington, Seattle, WA, United States
| | - Defu Zeng
- Irell and Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, United States.,Department of Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, CA, United States.,Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
20
|
Zitzer NC, Garzon R, Ranganathan P. Toll-Like Receptor Stimulation by MicroRNAs in Acute Graft-vs.-Host Disease. Front Immunol 2018; 9:2561. [PMID: 30455702 PMCID: PMC6230675 DOI: 10.3389/fimmu.2018.02561] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022] Open
Abstract
Acute graft-vs.-host disease (aGVHD) is a frequent complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT), accounting for substantial morbidity and mortality associated with this treatment modality. The pathogenesis of aGVHD involves a complex cascade of humoral and cellular interactions in which donor T cells target HLA mismatched host tissues, causing tissue injury through secretion of pro-inflammatory cytokines and induction of direct cytotoxicity. Toll-like receptors (TLRs) are key components of the innate immune system that recognize endogenous danger-associated molecular patterns (DAMPs) and exogenous pathogen-associated molecular patterns (PAMPs). Patients receiving conditioning chemotherapy and/or whole-body irradiation prior to all-HSCT are prone to gastrointestinal damage and translocation of microbiota across compromised intestinal epithelium, resulting in release of PAMPs and DAMPs. These “danger signals” play critical roles in disease pathogenesis by both initiating and propagating aGVHD through dendritic cell maturation and alloreactive T cell responses. There are 10–15 TLRs identified in mammalian species, a subset of which recognize single-stranded RNA (ssRNA) and serve as a key component of viral immunity. Recently, ssRNAs other than those of viral origin have been investigated as potential ligands of TLRs. MicroRNAs (miRs) are short (19–24 nt) non-coding RNAs that play critical roles in a variety of diseases. While traditionally miRs post-translationally modulate gene expression, non-canonical functions such as regulating TLR stimulation by acting as TLR ligands have been described. Here, we review the role of TLRs in aGVHD pathogenesis, the function of miRs in TLR stimulation, and the recent literature describing miRs as TLR ligands in aGVHD.
Collapse
Affiliation(s)
- Nina C Zitzer
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Ramiro Garzon
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Parvathi Ranganathan
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
21
|
Koenecke C, Krueger A. MicroRNA in T-Cell Development and T-Cell Mediated Acute Graft-Versus-Host Disease. Front Immunol 2018; 9:992. [PMID: 29867969 PMCID: PMC5949326 DOI: 10.3389/fimmu.2018.00992] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/20/2018] [Indexed: 12/21/2022] Open
Abstract
Acute graft-versus-host disease (GvHD) is still a major cause of treatment-related mortality after allogeneic stem cell transplantation. Allo-antigen recognition of donor T cells after transplantation account for the onset and persistence of this disease. MicroRNAs (miRNAs) are molecular regulators involved in numerous processes during T-cell development, homeostasis, and activation. Thus, miRNAs also contribute to pathological T-cell function during GvHD. Given their capacity of fine-tuning T-cell function, miRNAs have emerged as promising therapeutic targets to curtail acute GvHD, but simultaneously maintain T-cell-mediated graft-versus-tumor effects. Here, we review the role of key miRNAs contributing to the pathophysiology of GvHD. We focus on those miRNAs acting in T cells and for which a role in GvHD has been established in preclinical models. Finally, we provide an outlook for clinical application of this new therapeutic target for GvHD prevention and treatment.
Collapse
Affiliation(s)
- Christian Koenecke
- Clinic for Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Andreas Krueger
- Institute for Molecular Medicine, Goethe-University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
22
|
Sykes M. Immune monitoring of transplant patients in transient mixed chimerism tolerance trials. Hum Immunol 2018; 79:334-342. [PMID: 29289741 PMCID: PMC5924718 DOI: 10.1016/j.humimm.2017.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 12/31/2022]
Abstract
This review focuses on mechanistic studies performed in recipients of non-myeloablative bone marrow transplant regimens developed at Massachusetts General Hospital in HLA-identical and HLA-mismatched haploidentical combinations, initially as a platform for treatment of hematologic malignancies with immunotherapy in the form of donor leukocyte infusions, and later in combination with donor kidney transplantation for the induction of allograft tolerance. In patients with permanent mixed chimerism, central deletion may be a major mechanism of long-term tolerance. In patients in whom donor chimerism is only transient, the kidney itself plays a significant role in maintaining long-term tolerance. A high throughput sequencing approach to identifying and tracking a significant portion of the alloreactive T cell receptor repertoire has demonstrated biological significance in transplant patients and has been useful in pointing to clonal deletion as a long-term tolerance mechanism in recipients of HLA-mismatched combined kidney and bone marrow transplants with only transient chimerism.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Columbia University Medical Center, NY, USA; Department of Medicine, Columbia University Medical Center, NY, USA; Department of Microbiology & Immunology, Columbia University Medical Center, NY, USA; Department of Surgery, Columbia University Medical Center, NY, USA.
| |
Collapse
|
23
|
Al Malki MM, Jones R, Ma Q, Lee D, Reisner Y, Miller JS, Lang P, Hongeng S, Hari P, Strober S, Yu J, Maziarz R, Mavilio D, Roy DC, Bonini C, Champlin RE, Fuchs EJ, Ciurea SO. Proceedings From the Fourth Haploidentical Stem Cell Transplantation Symposium (HAPLO2016), San Diego, California, December 1, 2016. Biol Blood Marrow Transplant 2018; 24:895-908. [PMID: 29339270 PMCID: PMC7187910 DOI: 10.1016/j.bbmt.2018.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 02/04/2023]
Abstract
The resurgence of haploidentical stem cell transplantation (HaploSCT) over the last decade is one of the most important advances in the field of hematopoietic stem cell transplantation (HSCT). The modified platforms of T cell depletion either ex vivo (CD34+ cell selection, "megadoses" of purified CD34+ cells, or selective depletion of T cells) or newer platforms of in vivo depletion of T cells, with either post-transplantation high-dose cyclophosphamide or intensified immune suppression, have contributed to better outcomes, with survival similar to that in HLA-matched donor transplantation. Further efforts are underway to control viral reactivation using modified T cells, improve immunologic reconstitution, and decrease the relapse rate post-transplantation using donor-derived cellular therapy products, such as genetically modified donor lymphocytes and natural killer cells. Improvements in treatment-related mortality have allowed the extension of haploidentical donor transplants to patients with hemoglobinopathies, such as thalassemia and sickle cell disease, and the possible development of platforms for immunotherapy in solid tumors. Moreover, combining HSCT from a related donor with solid organ transplantation could allow early tapering of immunosuppression in recipients of solid organ transplants and hopefully prevent organ rejection in this setting. This symposium summarizes some of the most important recent advances in HaploSCT and provides a glimpse in the future of fast growing field.
Collapse
Affiliation(s)
- Monzr M Al Malki
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, California
| | - Richard Jones
- Division of Hematologic Malignancies, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, Maryland
| | - Qing Ma
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Dean Lee
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Yair Reisner
- Department of Immunology, Weizmann Institute, Rehovot, Israel
| | - Jeffrey S Miller
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Peter Lang
- Department of General Paediatrics, Oncology/Haematology, Tübingen University Hospital for Children and Adolescents, Tübingen, Germany
| | - Suradej Hongeng
- Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Parameswaran Hari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Samuel Strober
- Division of Immunology and Rheumatology, Department of Medicine, Stanford Medical School, Palo Alto, California
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Richard Maziarz
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Denis-Claude Roy
- Blood and Marrow Transplantation Program, Hôpital Maisonneuve-Rosemont Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Chiara Bonini
- Experimental Hematology Unit, San Raffaele Hospital, Milan, Italy
| | | | - Ephraim J Fuchs
- Division of Hematologic Malignancies, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, Maryland
| | - Stefan O Ciurea
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
24
|
Khan AB, Carpenter B, Santos e Sousa P, Pospori C, Khorshed R, Griffin J, Velica P, Zech M, Ghorashian S, Forrest C, Thomas S, Gonzalez Anton S, Ahmadi M, Holler A, Flutter B, Ramirez-Ortiz Z, Means TK, Bennett CL, Stauss H, Morris E, Lo Celso C, Chakraverty R. Redirection to the bone marrow improves T cell persistence and antitumor functions. J Clin Invest 2018; 128:2010-2024. [PMID: 29485974 PMCID: PMC5919805 DOI: 10.1172/jci97454] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/20/2018] [Indexed: 12/13/2022] Open
Abstract
A key predictor for the success of gene-modified T cell therapies for cancer is the persistence of transferred cells in the patient. The propensity of less differentiated memory T cells to expand and survive efficiently has therefore made them attractive candidates for clinical application. We hypothesized that redirecting T cells to specialized niches in the BM that support memory differentiation would confer increased therapeutic efficacy. We show that overexpression of chemokine receptor CXCR4 in CD8+ T cells (TCXCR4) enhanced their migration toward vascular-associated CXCL12+ cells in the BM and increased their local engraftment. Increased access of TCXCR4 to the BM microenvironment induced IL-15-dependent homeostatic expansion and promoted the differentiation of memory precursor-like cells with low expression of programmed death-1, resistance to apoptosis, and a heightened capacity to generate polyfunctional cytokine-producing effector cells. Following transfer to lymphoma-bearing mice, TCXCR4 showed a greater capacity for effector expansion and better tumor protection, the latter being independent of changes in trafficking to the tumor bed or local out-competition of regulatory T cells. Thus, redirected homing of T cells to the BM confers increased memory differentiation and antitumor immunity, suggesting an innovative solution to increase the persistence and functions of therapeutic T cells.
Collapse
Affiliation(s)
- Anjum B. Khan
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Ben Carpenter
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Pedro Santos e Sousa
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Constandina Pospori
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Reema Khorshed
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - James Griffin
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Pedro Velica
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Mathias Zech
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Sara Ghorashian
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Calum Forrest
- University College London (UCL) Cancer Institute, London, United Kingdom
| | - Sharyn Thomas
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Sara Gonzalez Anton
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Maryam Ahmadi
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Angelika Holler
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Barry Flutter
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Zaida Ramirez-Ortiz
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Terry K. Means
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Clare L. Bennett
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Hans Stauss
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Emma Morris
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Cristina Lo Celso
- Department of Life Sciences, Imperial College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Ronjon Chakraverty
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| |
Collapse
|
25
|
Dierckx de Casterlé I, Billiau AD, Sprangers B. Recipient and donor cells in the graft-versus-solid tumor effect: It takes two to tango. Blood Rev 2018; 32:449-456. [PMID: 29678553 DOI: 10.1016/j.blre.2018.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/05/2018] [Accepted: 04/06/2018] [Indexed: 12/16/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (alloHSCT) produces -similar to the long-established graft-versus-leukemia effect- graft-versus-solid-tumor effects. Clinical trials reported response rates of up to 53%, occurring mostly but not invariably in association with full donor chimerism and/or graft-versus-host disease. Although donor-derived T cells are considered the principal effectors of anti-tumor immunity after alloHSCT or donor leukocyte infusion (DLI), growing evidence indicate that recipient-derived immune cells may also contribute. Whereas the role of recipient-derived antigen-presenting cells in eliciting graft-versus-host reactions and priming donor T cells following DLI is well known, resulting inflammatory responses may also break tolerance of recipient effector cells towards the tumor. Additionally, mouse studies indicated that post-transplant recipient leukocyte infusion produces anti-leukemia and anti-solid-tumor effects that were exclusively mediated by recipient-type effector cells, without graft-versus-host disease. Here, we review current preclinical and clinical evidence on graft-versus-solid-tumor effects and growing evidence on the effector role of recipient-derived immune cells in the anti-tumor effect of alloHSCT.
Collapse
Affiliation(s)
- Isabelle Dierckx de Casterlé
- Department of Microbiology and Immunology, Laboratory of Experimental Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - An D Billiau
- Department of Microbiology and Immunology, Laboratory of Experimental Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - Ben Sprangers
- Department of Microbiology and Immunology, Laboratory of Experimental Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Nephrology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
26
|
Santos e Sousa P, Ciré S, Conlan T, Jardine L, Tkacz C, Ferrer IR, Lomas C, Ward S, West H, Dertschnig S, Blobner S, Means TK, Henderson S, Kaplan DH, Collin M, Plagnol V, Bennett CL, Chakraverty R. Peripheral tissues reprogram CD8+ T cells for pathogenicity during graft-versus-host disease. JCI Insight 2018; 3:97011. [PMID: 29515032 PMCID: PMC5922296 DOI: 10.1172/jci.insight.97011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 02/07/2018] [Indexed: 01/05/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic stem cell transplantation induced by the influx of donor-derived effector T cells (TE) into peripheral tissues. Current treatment strategies rely on targeting systemic T cells; however, the precise location and nature of instructions that program TE to become pathogenic and trigger injury are unknown. We therefore used weighted gene coexpression network analysis to construct an unbiased spatial map of TE differentiation during the evolution of GVHD and identified wide variation in effector programs in mice and humans according to location. Idiosyncrasy of effector programming in affected organs did not result from variation in T cell receptor repertoire or the selection of optimally activated TE. Instead, TE were reprogrammed by tissue-autonomous mechanisms in target organs for site-specific proinflammatory functions that were highly divergent from those primed in lymph nodes. In the skin, we combined the correlation-based network with a module-based differential expression analysis and showed that Langerhans cells provided in situ instructions for a Notch-dependent T cell gene cluster critical for triggering local injury. Thus, the principal determinant of TE pathogenicity in GVHD is the final destination, highlighting the need for target organ-specific approaches to block immunopathology while avoiding global immune suppression.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Bone Marrow Transplantation/adverse effects
- Cells, Cultured
- Cellular Reprogramming/genetics
- Cellular Reprogramming/immunology
- Disease Models, Animal
- Female
- Gene Expression Regulation/immunology
- Graft vs Host Disease/immunology
- Graft vs Host Disease/pathology
- Hematopoietic Stem Cell Transplantation/adverse effects
- Humans
- Langerhans Cells/immunology
- Langerhans Cells/metabolism
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Male
- Mannose-Binding Lectins/genetics
- Mannose-Binding Lectins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Multigene Family/genetics
- Multigene Family/immunology
- Primary Cell Culture
- Receptors, Notch/metabolism
- Skin/cytology
- Skin/immunology
- Skin/pathology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Transplantation Chimera
- Transplantation, Homologous/adverse effects
Collapse
Affiliation(s)
- Pedro Santos e Sousa
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Séverine Ciré
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Thomas Conlan
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Laura Jardine
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | | - Ivana R. Ferrer
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Cara Lomas
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Sophie Ward
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Heather West
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Simone Dertschnig
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Sven Blobner
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Terry K. Means
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | | | - Daniel H. Kaplan
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew Collin
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | | - Clare L. Bennett
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| | - Ronjon Chakraverty
- Haematology, UCL Cancer Institute and Institute of Immunity & Transplantation, London, United Kingdom (UK)
| |
Collapse
|
27
|
Abstract
Acute graft-versus-host disease (GVHD) in the gut is common following hematopoetic cell transplantation (HCT) and is associated with high mortality. However, it remains unclear whether Th1 or Th17 CD4+ T cells can initiate acute gut GVHD. In this issue of the JCI, Ullrich and colleagues identified a subset of CD4+ T cells that express high levels of IL-7Rα and granulocyte-macrophage CSF (IL-7RαhiGM-CSF+) cells that are involved in the induction of acute gut GVHD in murine models. The IL-7RαhiGM-CSF+ effector memory cells were BATF dependent, RORγt independent, produced large amounts of GM-CSF and IFN-γ, and released little IL-17. CD4+IL-7RαhiGM-CSF+ cells were not classical Th17 cells but had more of a Th1-like phenotype, despite their dependence on BATF. This work suggests that targeting the IL-7R/BATF/GM-CSF axis has therapeutic potential for treating acute gut GVHD.
Collapse
|
28
|
West HC, Bennett CL. Redefining the Role of Langerhans Cells As Immune Regulators within the Skin. Front Immunol 2018; 8:1941. [PMID: 29379502 PMCID: PMC5770803 DOI: 10.3389/fimmu.2017.01941] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/18/2017] [Indexed: 12/28/2022] Open
Abstract
Langerhans cells (LC) are a unique population of tissue-resident macrophages that form a network of cells across the epidermis of the skin, but which have the ability to migrate from the epidermis to draining lymph nodes (LN). Their location at the skin barrier suggests a key role as immune sentinels. However, despite decades of research, the role of LC in skin immunity is unclear; ablation of LC results in neither fatal susceptibility to skin infection nor overt autoimmunity due to lack of immune regulation. Our understanding of immune processes has traditionally been centered on secondary lymphoid organs as sites of lymphocyte priming and differentiation, which is exemplified by LC, initially defined as a paradigm for tissue dendritic cells that migrate to draining LN on maturation. But, more recently, an awareness of the importance of the tissue environment in shaping effector immunity has emerged. In this mini-review, we discuss whether our lack of understanding of LC function stems from our lymph node-centric view of these cells, and question whether a focus on LC as immune regulators in situ in the skin may reveal clearer answers about their function in cutaneous immunology.
Collapse
Affiliation(s)
- Heather C. West
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
- Division of Cancer Studies, University College London, London, United Kingdom
| | - Clare L. Bennett
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
- Division of Cancer Studies, University College London, London, United Kingdom
| |
Collapse
|
29
|
Reddy P, Ferrara JL. Graft-Versus-Host Disease and Graft-Versus-Leukemia Responses. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
30
|
Zuber J, Sykes M. Mechanisms of Mixed Chimerism-Based Transplant Tolerance. Trends Immunol 2017; 38:829-843. [PMID: 28826941 PMCID: PMC5669809 DOI: 10.1016/j.it.2017.07.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/24/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023]
Abstract
Immune responses to allografts represent a major barrier in organ transplantation. Immune tolerance to avoid chronic immunosuppression is a critical goal in the field, recently achieved in the clinic by combining bone marrow transplantation (BMT) with kidney transplantation following non-myeloablative conditioning. At high levels of chimerism such protocols can permit central deletional tolerance, but with a significant risk of graft-versus-host (GVH) disease (GVHD). By contrast, transient chimerism-based tolerance is devoid of GVHD risk and appears to initially depend on regulatory T cells (Tregs) followed by gradual, presumably peripheral, clonal deletion of donor-reactive T cells. Here we review recent mechanistic insights into tolerance and the development of more robust and safer protocols for tolerance induction that will be guided by innovative immune monitoring tools.
Collapse
Affiliation(s)
- Julien Zuber
- Service de Transplantation Rénale, Hôpital Necker, Université Paris Descartes, Paris, France; INSERM UMRS_1163, IHU Imagine, Paris, France.
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Surgery, Columbia University, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Center, New York, NY 10032, USA.
| |
Collapse
|
31
|
Isolated Extramedullary Relapse of Acute Leukemia after Allogeneic Stem Cell Transplantation: Different Kinetics and Better Prognosis than Systemic Relapse. Biol Blood Marrow Transplant 2017; 23:1087-1094. [DOI: 10.1016/j.bbmt.2017.03.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/20/2017] [Indexed: 11/19/2022]
|
32
|
Dodge J, Stephans A, Lai J, Drobyski WR, Chen X. Effects of Donor Vitamin A Deficiency and Pharmacologic Modulation of Donor T Cell Retinoic Acid Pathway on the Severity of Experimental Graft-versus-Host Disease. Biol Blood Marrow Transplant 2016; 22:2141-2148. [PMID: 27596131 DOI: 10.1016/j.bbmt.2016.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/01/2016] [Indexed: 02/08/2023]
Abstract
Graft-versus-host disease (GVHD) is the major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). A combination of genetic and nongenetic factors dictates the incidence and severity of GVHD. Recent studies have identified the potential role of the retinoic acid (RA)/retinoic acid receptor (RAR) pathway in the pathogenesis of GVHD. RA is the active metabolite of vitamin A. Thus, a clinically relevant question is whether HSCT donor and/or recipient vitamin A status affects the development of GVHD. It has been previously reported that recipient vitamin A deficiency is associated with reduced intestinal GVHD and prolonged overall survival after experimental allogeneic HSCT. However, it is still unknown whether donor vitamin A status influences GVHD development. In the current study, we report that chronic vitamin A deficiency changes the composition of T cell compartment of donor mice with a reduction in the percentage of CD4+ T cells. We showed that although vitamin A deficiency does not affect donor T cell alloreactivity on a per cell basis, a decreased proportion of donor CD4+ T cells in marrow graft inoculums leads to reduced incidence and severity of GVHD. Furthermore, our proof of principle studies using a pan-RAR antagonist demonstrated that transient inhibition of donor T cell RAR signaling can reduce T cell alloreactivity and their ability to cause lethal GVHD. Our studies provide preclinical evidence that donor vitamin A deficiency may be a nongenetic factor that can modulate the severity of GVHD and pharmacologic interfering RA/RAR pathway in donor T cells might be a valuable approach for mitigating GVHD after allogeneic HSCT.
Collapse
Affiliation(s)
- Joseph Dodge
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allison Stephans
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jinping Lai
- Department of Pathology, St. Louis University School of Medicine, St. Louis, Missouri
| | - William R Drobyski
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiao Chen
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
33
|
Manzo T, Sturmheit T, Basso V, Petrozziello E, Hess Michelini R, Riba M, Freschi M, Elia AR, Grioni M, Curnis F, Protti MP, Schumacher TN, Debets R, Swartz MA, Corti A, Bellone M, Mondino A. T Cells Redirected to a Minor Histocompatibility Antigen Instruct Intratumoral TNFα Expression and Empower Adoptive Cell Therapy for Solid Tumors. Cancer Res 2016; 77:658-671. [PMID: 27872095 DOI: 10.1158/0008-5472.can-16-0725] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 10/31/2016] [Accepted: 11/09/2016] [Indexed: 11/16/2022]
Abstract
Donor-derived allogeneic T cells evoke potent graft versus tumor (GVT) effects likely due to the simultaneous recognition of tumor-specific and host-restricted minor histocompatibility (H) antigens. Here we investigated whether such effects could be reproduced in autologous settings by TCR gene-engineered lymphocytes. We report that T cells redirected either to a broadly expressed Y-encoded minor H antigen or to a tumor-associated antigen, although poorly effective if individually transferred, when simultaneously administered enabled acute autochthonous tumor debulking and resulted in durable clinical remission. Y-redirected T cells proved hyporesponsive in peripheral lymphoid organs, whereas they retained effector function at the tumor site, where in synergy with tumor-redirected lymphocytes, they instructed TNFα expression, endothelial cell activation, and intratumoral T-cell infiltration. While neutralizing TNFα hindered GVT effects by the combined T-cell infusion, a single injection of picogram amounts of NGR-TNF, a tumor vessel-targeted TNFα derivative currently in phase III clinical trials, substituted for Y-redirected cells and enabled tumor debulking by tumor-redirected lymphocytes. Together, our results provide new mechanistic insights into allogeneic GVT, validate the importance of targeting the tumor and its associated stroma, and prove the potency of a novel combined approach suitable for immediate clinical implementation. Cancer Res; 77(3); 658-71. ©2016 AACR.
Collapse
Affiliation(s)
- Teresa Manzo
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | - Tabea Sturmheit
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | - Veronica Basso
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Petrozziello
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | - Rodrigo Hess Michelini
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy
| | - Michela Riba
- Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Freschi
- Department of Pathology, San Raffaele Scientific Institute, Milan, Italy
| | - Angela R Elia
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Grioni
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy
| | - Flavio Curnis
- Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Protti
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy
| | - Ton N Schumacher
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Melody A Swartz
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Institute for Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Angelo Corti
- Università Vita-Salute San Raffaele, Milan, Italy.,Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Bellone
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Mondino
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
34
|
Conditioning with Fludarabine-Busulfan versus Busulfan-Cyclophosphamide Is Associated with Lower aGVHD and Higher Survival but More Extensive and Long Standing Bone Marrow Damage. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3071214. [PMID: 27843940 PMCID: PMC5098055 DOI: 10.1155/2016/3071214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/15/2016] [Accepted: 09/18/2016] [Indexed: 11/18/2022]
Abstract
Acute graft-versus-host disease (aGVHD) is a major complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT) and a major cause of nonrelapse mortality after allo-HSCT. A conditioning regimen plays a pivotal role in the development of aGVHD. To provide a platform for studying aGVHD and evaluating the impact of different conditioning regimens, we established a murine aGVHD model that simulates the clinical situation and can be conditioned with Busulfan-Cyclophosphamide (Bu-Cy) and Fludarabine-Busulfan (Flu-Bu). In our study, BALB/c mice were conditioned with Bu-Cy or Flu-Bu and transplanted with 2 × 107 bone marrow cells and 2 × 107 splenocytes from either allogeneic (C57BL/6) or syngeneic (BALB/c) donors. The allogeneic recipients conditioned with Bu-Cy had shorter survivals (P < 0.05), more severe clinical manifestations, and higher hepatic and intestinal pathology scores, associated with increased INF-γ expression and diminished IL-4 expression in serum, compared to allogeneic recipients conditioned with Flu-Bu. Moreover, higher donor-derived T-cell infiltration and severely impaired B-cell development were seen in the bone marrow of mice, exhibiting aGVHD and conditioned with Flu-Bu. Our study showed that the conditioning regimen with Bu-Cy resulted in more severe aGVHD while the Flu-Bu regimen was associated with more extensive and long standing bone marrow damage.
Collapse
|
35
|
Hu R, Liu Y, Song Y, Su M, Lu X, Rood D, Lai L. Recombinant IL-7/HGFβ hybrid cytokine separates acute graft-versus-host-disease from graft-versus-tumour activity by altering donor T cell trafficking. Br J Haematol 2016; 175:505-516. [PMID: 27447780 DOI: 10.1111/bjh.14268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/07/2016] [Indexed: 11/29/2022]
Abstract
Given that donor T cells from a transplant contribute both the desired graft-versus-tumour (GVT) effect and detrimental graft-versus-host disease (GVHD), strategies to separate GVHD and GVT activity are a major clinical goal. We have previously demonstrated that in vivo administration of a recombinant (r)IL-7/HGFβ hybrid cytokine, consisting of interleukin-7 (IL-7, IL7) and the β-chain of hepatocyte growth factor (HGFβ), significantly inhibits the growth of cancer cells in murine tumour models. The antit-umour effect of rIL-7/HGFβ is related to a marked infiltration T cells in the tumour tissues. We have also shown that GVHD was not induced in rIL-7/HGFβ-treated T cell-depleted allogeneic haematopoietic stem cell transplantation (HSCT) recipients. We show here that, in T cell-replete allogeneic HSCT murine models, rIL-7/HGFβ attenuated acute GVHD (aGVHD), while promoting GVT activity. This was related to an alteration of donor T cell trafficking, with an increased infiltration of donor T cells into tumour tissues and the lympho-haematopoietic system but decreased number of the T cells in the GVHD target organs. Therefore, rIL-7/HGFβ may offer a new tool to alleviate aGVHD while prompting GVT, and to study the molecular regulation of T cell trafficking.
Collapse
Affiliation(s)
- Rong Hu
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA.,Guizhou Medical University, Guizhou, China
| | - Yalan Liu
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Yinhong Song
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Xiuling Lu
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Debra Rood
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA. .,University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
36
|
Watts JM, Wang XV, Swords RT, Paietta E, Douer D, Lugar SM, Fernandez HF, Rowe JM, Lazarus HM, Tallman MS, Litzow MR. Very late relapse of AML after allogeneic hematopoietic cell transplantation is often extramedullary. Bone Marrow Transplant 2016; 51:1013-5. [PMID: 26974275 DOI: 10.1038/bmt.2016.44] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- J M Watts
- Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - X V Wang
- Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard School of Public Health, Boston, MA, USA
| | - R T Swords
- Miller School of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - E Paietta
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - D Douer
- Leukemia Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - S M Lugar
- University of Pennsylvania, Philadelphia, PA, USA
| | - H F Fernandez
- H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - J M Rowe
- Shaare Zedek Medical Center, Jerusalem, Israel
| | - H M Lazarus
- Case Western Reserve University, Cleveland, OH, USA
| | - M S Tallman
- Leukemia Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | | |
Collapse
|
37
|
Potential protective effect of Helicobacter pylori on the development of gastrointestinal GvHD. Bone Marrow Transplant 2016; 51:819-24. [PMID: 26950379 DOI: 10.1038/bmt.2016.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 01/21/2016] [Accepted: 01/24/2016] [Indexed: 11/08/2022]
Abstract
Previous reports ascribe a modulating capacity of the immune response to Helicobacter pylori (HP). Our hypothesis was to demonstrate in a prospective study that HP infection could have a protective effect against development of gastrointestinal GvHD in patients receiving allogeneic hematopoietic cell transplantation (HCT). Presence of HP before transplant was determined using C(13) urea breath test. Seventy-nine patients receiving an allogeneic HCT were included and 93.7% of them received PBSC; in 51.9%, the donor was unrelated. Acute gastrointestinal GvHD was diagnosed in 51.9% (n=41). In the multivariable analysis, HP infection was associated with a lower frequency of gastrointestinal GvHD (odds ratio (OR)=0.19 (95% confidence interval (CI): 0.05-0.67); in contrast, an unrelated donor was associated with a higher frequency of gastrointestinal GvHD (odds ratio=5.4 (95% confidence interval: 1.6-18.2). One year overall survival (OS) was 74%. In the multivariate Cox proportional-hazards regression analysis, stages 0-II gastrointestinal GvHD (hazards ratio (HR)=0.19), reduced intensity conditioning (HR=0.04) and tacrolimus-sirolimus GvHD prophylaxis (HR=0.06) were all associated with a better OS. In summary, HP infection could have a role in decreasing gastrointestinal GvHD in patients receiving allogeneic HCT from peripheral blood including related and unrelated donors.
Collapse
|
38
|
Sykes M. Immune tolerance in recipients of combined haploidentical bone marrow and kidney transplantation. Bone Marrow Transplant 2016; 50 Suppl 2:S82-6. [PMID: 26039215 DOI: 10.1038/bmt.2015.102] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The success of allogeneic hematopoietic cell transplantation (HCT) has been limited by transplant-associated toxicities related to the conditioning regimens used and to graft-vs-host disease (GVHD). The frequency and severity of GVHD observed when extensive HLA barriers are transgressed has greatly impeded the routine use of extensively HLA-mismatched HCT. Allogeneic HCT also has potential as an approach to organ allograft tolerance induction, but this potential has not been previously realized because of the toxicity associated with traditional conditioning. This paper reviews an approach to HCT involving reduced intensity conditioning that demonstrated sufficient safety in patients with hematologic malignancies, even in the HLA-mismatched transplant setting, to be applied for the induction of kidney allograft tolerance in humans with no other indication for HCT. These studies provided the first successful example of intentional organ allograft tolerance induction across HLA barriers in humans. Current data and hypotheses on the mechanisms of tolerance in these patients are reviewed.
Collapse
Affiliation(s)
- M Sykes
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
39
|
Michonneau D, Sagoo P, Breart B, Garcia Z, Celli S, Bousso P. The PD-1 Axis Enforces an Anatomical Segregation of CTL Activity that Creates Tumor Niches after Allogeneic Hematopoietic Stem Cell Transplantation. Immunity 2016; 44:143-154. [DOI: 10.1016/j.immuni.2015.12.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/18/2015] [Accepted: 10/12/2015] [Indexed: 12/21/2022]
|
40
|
Li HW, Andreola G, Carlson AL, Shao S, Lin CP, Zhao G, Sykes M. Rapid Functional Decline of Activated and Memory Graft-versus-Host-Reactive T Cells Encountering Host Antigens in the Absence of Inflammation. THE JOURNAL OF IMMUNOLOGY 2015; 195:1282-92. [PMID: 26085679 DOI: 10.4049/jimmunol.1401511] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 05/22/2015] [Indexed: 01/04/2023]
Abstract
Inflammation in the priming host environment has critical effects on the graft-versus-host (GVH) responses mediated by naive donor T cells. However, it is unclear how a quiescent or inflammatory environment impacts the activity of GVH-reactive primed T and memory cells. We show in this article that GVH-reactive primed donor T cells generated in irradiated recipients had diminished ability compared with naive T cells to increase donor chimerism when transferred to quiescent mixed allogeneic chimeras. GVH-reactive primed T cells showed marked loss of cytotoxic function and activation, and delayed but not decreased proliferation or accumulation in lymphoid tissues when transferred to quiescent mixed chimeras compared with freshly irradiated secondary recipients. Primed CD4 and CD8 T cells provided mutual help to sustain these functions in both subsets. CD8 help for CD4 cells was largely IFN-γ dependent. TLR stimulation after transfer of GVH-reactive primed T cells to mixed chimeras restored their cytotoxic effector function and permitted the generation of more effective T cell memory in association with reduced PD-1 expression on CD4 memory cells. Our data indicate that an inflammatory host environment is required for the maintenance of GVH-reactive primed T cell functions and the generation of memory T cells that can rapidly acquire effector functions. These findings have important implications for graft-versus-host disease and T cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Hao Wei Li
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Giovanna Andreola
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Alicia L Carlson
- Advanced Microscopy Program, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114; and Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Steven Shao
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032
| | - Charles P Lin
- Advanced Microscopy Program, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114; and Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Guiling Zhao
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Megan Sykes
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129;
| |
Collapse
|
41
|
Okiyama N, Fujimoto M. Clinical perspectives and murine models of lichenoid tissue reaction/interface dermatitis. J Dermatol Sci 2015; 78:167-72. [PMID: 25813248 DOI: 10.1016/j.jdermsci.2015.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 02/26/2015] [Accepted: 03/02/2015] [Indexed: 01/05/2023]
Abstract
A set of histopathological elements, that is death of epidermal basal cell layer keratinocytes and inflammatory cell infiltration, distinguishes lichenoid tissue reaction (LTR)/interface dermatitis (IFD) from other inflammatory mucocutaneous diseases with histological findings of superficial perivascular dermatitis. The LTR/IFD is observed in inflammatory mucocutaneous diseases such as lichen planus, Stevens-Johnson syndrome/toxic epidermal necrolysis, acute graft-versus-host disease, lupus erythematosus and dermatomyositis. Clinical and basic researches have suggested that keratinocytes are antigen-presenting cells and mediate LTR/IFD reaction via production of cytokines/chemokines and inhibitory molecules such as programmed cell death (PD)-L1, and that cytotoxic CD8(+) T cells producing cytotoxic granules, perforin, granzyme B and granulysin are final effector cells to cause keratinocyte death. Because interferon-γ and FasL, which are produced by not only CD8(+) but also CD4(+) T cells, are candidates of the pathogenic molecules in LTR/IFD, CD4(+) T cells may also play a role to develop LTR/IFD. On the other hand, CD4(+) Treg cells accelerate the remission of LTR/IFD. Some murine models of LTR/IFD have been established. For example, LTR/IFD reactions were induced in keratinocyte-specific membrane-binding ovalbumin-transgenic (mOVA Tg) mice by adoptive transfer of CD8(+) T cells with OVA-specific T-cell-receptor. It has also been shown that human CD8(+) T cells are pathogenic immune cells in human skin-xenografted mice. Various immunosuppressants are used to treat patients with mucocutaneous diseases with LTR/IFD. By analysis of the mOVA Tg mice, a JAK inhibitor was suggested to be a new candidate drug to inhibit not only pathogenic T cells but also keratinocyte death in LTR/IFD. More specific treatments for patients with LTR/IFD will be developed in future.
Collapse
Affiliation(s)
- Naoko Okiyama
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan.
| | - Manabu Fujimoto
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| |
Collapse
|
42
|
Sporrer D, Gessner A, Hehlgans T, Oefner PJ, Holler E. The Microbiome and Allogeneic Stem Cell Transplantation. CURRENT STEM CELL REPORTS 2015. [DOI: 10.1007/s40778-014-0006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
43
|
Nalle SC, Kwak HA, Edelblum KL, Joseph NE, Singh G, Khramtsova GF, Mortenson ED, Savage PA, Turner JR. Recipient NK cell inactivation and intestinal barrier loss are required for MHC-matched graft-versus-host disease. Sci Transl Med 2015; 6:243ra87. [PMID: 24990882 DOI: 10.1126/scitranslmed.3008941] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have shown a correlation between pretransplant conditioning intensity, intestinal barrier loss, and graft-versus-host disease (GVHD) severity. However, because irradiation and other forms of pretransplant conditioning have pleiotropic effects, the precise role of intestinal barrier loss in GVHD pathogenesis remains unclear. We developed GVHD models that allowed us to isolate the specific contributions of distinct pretransplant variables. Intestinal damage was required for the induction of minor mismatch [major histocompatibility complex (MHC)-matched] GVHD, but was not necessary for major mismatch GVHD, demonstrating fundamental pathogenic distinctions between these forms of disease. Moreover, recipient natural killer (NK) cells prevented minor mismatch GVHD by limiting expansion and target organ infiltration of alloreactive T cells via a perforin-dependent mechanism, revealing an immunoregulatory function of MHC-matched recipient NK cells in GVHD. Minor mismatch GVHD required MyD88-mediated Toll-like receptor 4 (TLR4) signaling on donor cells, and intestinal damage could be bypassed by parenteral lipopolysaccharide (LPS) administration, indicating a critical role for the influx of bacterial components triggered by intestinal barrier loss. In all, the data demonstrate that pretransplant conditioning plays a dual role in promoting minor mismatch GVHD by both depleting recipient NK cells and inducing intestinal barrier loss.
Collapse
Affiliation(s)
- Sam C Nalle
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - H Aimee Kwak
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Karen L Edelblum
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Nora E Joseph
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Gurminder Singh
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | | | - Eric D Mortenson
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Peter A Savage
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| | - Jerrold R Turner
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
44
|
Wang H, Yang YG. The complex and central role of interferon-γ in graft-versus-host disease and graft-versus-tumor activity. Immunol Rev 2015; 258:30-44. [PMID: 24517424 DOI: 10.1111/imr.12151] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 11/01/2013] [Accepted: 11/11/2013] [Indexed: 12/22/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is increasingly being performed to treat patients with hematologic malignancies. However, separating the beneficial graft-versus-tumor (GVT) or graft-versus-leukemia effects from graft-versus-host disease (GVHD) has been difficult and remains a significant challenge toward improving therapeutic efficacy and reducing toxicity of allo-HCT. GVHD is induced by donor T cells that also mediate potent anti-tumor responses. However, despite the largely shared effector mechanisms, extensive animal studies have demonstrated the potential of dissociating the GVT effect from GVHD. Also in many clinical cases, long-term remission was achieved following allo-HCT, without significant GVHD. A better mechanistic understanding of the immunopathophysiology of GVHD and GVT effects may potentially help to improve allo-HCT as well as maximize the benefit of GVT effects while minimizing GVHD. In this article, we review the role of IFN-γ in regulation of alloresponses following allo-HCT, with a focus on the mechanisms of how this cytokine may separate GVHD from GVT effects.
Collapse
Affiliation(s)
- Hui Wang
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | |
Collapse
|
45
|
Abstract
Vertebrates mount strong adaptive immune responses to transplanted organs (allografts), but the mechanisms by which the innate immune system initiates this response are not completely understood. In anti-microbial immunity, non-self molecules associated with pathogens but not present in the host induce the maturation of innate antigen-presenting cells (APCs) by binding to germ-line-encoded receptors. Mature APCs then initiate the adaptive immune response by presenting microbial antigen and providing costimulatory signals to T cells. How allografts activate APCs, however, is less clear, because allografts are presumably sterile. A widely accepted view is that inflammatory or 'danger' molecules released by dying graft cells at the time of transplantation trigger APC maturation and the T-cell response that follows. Alternatively, it has been proposed that the introduction of microbial products during the surgical procedure could also alert the innate immune system to the presence of the transplanted organ. Here, we review why these hypotheses fail to fully explain how the alloimmune response is initiated after transplantation and summarize evidence that recognition of allogeneic non-self by monocytes is a key event in triggering alloimmunity and graft rejection.
Collapse
Affiliation(s)
- Martin H Oberbarnscheidt
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine & University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh School of Medicine & University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | |
Collapse
|
46
|
Intravital Multiphoton Imaging of Cutaneous Immune Responses. J Invest Dermatol 2014; 134:2680-2684. [DOI: 10.1038/jid.2014.225] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/15/2014] [Accepted: 04/29/2014] [Indexed: 12/31/2022]
|
47
|
Hock K, Pilat N, Baranyi U, Mahr B, Gattringer M, Klaus C, Wekerle T. Donor CD4 T cells trigger costimulation blockade-resistant donor bone marrow rejection through bystander activation requiring IL-6. Am J Transplant 2014; 14:2011-22. [PMID: 25100658 DOI: 10.1111/ajt.12823] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 04/22/2014] [Accepted: 05/07/2014] [Indexed: 01/25/2023]
Abstract
Bone marrow (BM) transplantation under costimulation blockade induces chimerism and tolerance. Cotransplantation of donor T cells (contained in substantial numbers in mobilized peripheral blood stem cells and donor lymphocyte infusions) together with donor BM paradoxically triggers rejection of donor BM through undefined mechanisms. Here, nonmyeloablatively irradiated C57BL/6 recipients simultaneously received donor BM (BALB/c) and donor T cells under costimulation blockade (anti-CD154 and CTLA4Ig). Donor CD4, but not CD8 cells, triggered natural killer-independent donor BM rejection which was associated with increased production of IL-6, interferon gamma (IFN-γ) and IL-17A. BM rejection was prevented through neutralization of IL-6, but not of IFN-γ or IL-17A. IL-6 counteracted the antiproliferative effect of anti-CD154 in vitro. Rapamycin and anti-lymphocyte function-associated antigen 1 negated this effect of IL-6 in vitro and prevented BM rejection in vivo. Simultaneous cotransplantation of (BALB/cxB6)F1, recipient or irradiated donor CD4 cells, or late transfer of donor CD4 cells did not lead to BM rejection, whereas cotransplantation of third party CD4 cells did. Transferred donor CD4 cells became activated, rapidly underwent apoptosis and triggered activation and proliferation of recipient T cells. Collectively, these results provide evidence that donor T cells recognizing the recipient as allogeneic lead to the release of IL-6, which abolishes the effect of anti-CD154, triggering donor BM rejection through bystander activation.
Collapse
Affiliation(s)
- K Hock
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
48
|
Ghosh A, Holland AM, van den Brink MRM. Genetically engineered donor T cells to optimize graft-versus-tumor effects across MHC barriers. Immunol Rev 2014; 257:226-36. [PMID: 24329800 DOI: 10.1111/imr.12142] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Hematopoietic stem cell transplantation has been used for more than 50 years to combat hematologic malignancies. In addition to being the first stem cell therapy, transplantation has provided evidence for the potent anti-tumor effects of T cells. Facilitating T-cell-based immunity against malignancies requires a careful balancing act between generating a robust response and avoiding off-target killing of healthy tissues, which is difficult to accomplish using bulk donor T cells. To address these issues, several approaches have been developed, drawing on basic T-cell biology, to potentiate graft-versus-tumor activity while avoiding graft-versus-host disease. Current strategies for anti-tumor cell therapies include: (i) selecting optimal T cells for transfer; (ii) engineering T cells to possess enhanced effector functions; and (iii) generating T-cell precursors that complete development after adoptive transfer. In this review, we assess the current state of the art in T-lineage cell therapy to treat malignancies in the context of allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Arnab Ghosh
- Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | |
Collapse
|
49
|
Heidegger S, van den Brink MRM, Haas T, Poeck H. The role of pattern-recognition receptors in graft-versus-host disease and graft-versus-leukemia after allogeneic stem cell transplantation. Front Immunol 2014; 5:337. [PMID: 25101080 PMCID: PMC4102927 DOI: 10.3389/fimmu.2014.00337] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 07/03/2014] [Indexed: 02/04/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only treatment with curative potential for certain aggressive hematopoietic malignancies. Its success is limited by acute graft-versus-host disease (GVHD), a life-threatening complication that occurs when allo-reactive donor T cells attack recipient organs. There is growing evidence that microbes and innate pattern-recognition receptors (PRRs) such as toll-like receptors (TLR) and nod-like receptors (NLR) are critically involved in the pathogenesis of acute GVHD. Currently, a widely accepted model postulates that intensive chemotherapy and/or total-body irradiation during pre-transplant conditioning results in tissue damage and a loss of epithelial barrier function. Subsequent translocation of bacterial components as well as release of endogenous danger molecules stimulate PRRs of host antigen-presenting cells to trigger the production of pro-inflammatory cytokines (cytokine storm) that modulate T cell allo-reactivity against host tissues, but eventually also the beneficial graft-versus-leukemia (GVL) effect. Given the limitations of existing immunosuppressive therapies, a better understanding of the molecular mechanisms that govern GVHD versus GVL is urgently needed. This may ultimately allow to design modulators, which protect from GvHD but preserve donor T-cell attack on hematologic malignancies. Here, we will briefly summarize current knowledge about the role of innate immunity in the pathogenesis of GVHD and GVL following allo-HSCT.
Collapse
Affiliation(s)
- Simon Heidegger
- III. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München , Munich , Germany
| | - Marcel R M van den Brink
- Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center , New York, NY , USA
| | - Tobias Haas
- III. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München , Munich , Germany
| | - Hendrik Poeck
- III. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München , Munich , Germany ; Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center , New York, NY , USA
| |
Collapse
|
50
|
Abstract
Over the past 5 years, many novel approaches to early diagnosis, prevention, and treatment of acute graft-versus-host disease (aGVHD) have been translated from the bench to the bedside. In this review, we highlight recent discoveries in the context of current aGVHD care. The most significant innovations that have already reached the clinic are prophylaxis strategies based upon a refinement of our understanding of key sensors, effectors, suppressors of the immune alloreactive response, and the resultant tissue damage from the aGVHD inflammatory cascade. In the near future, aGVHD prevention and treatment will likely involve multiple modalities, including small molecules regulating immunologic checkpoints, enhancement of suppressor cytokines and cellular subsets, modulation of the microbiota, graft manipulation, and other donor-based prophylaxis strategies. Despite long-term efforts, major challenges in treatment of established aGVHD still remain. Resolution of inflammation and facilitation of rapid immune reconstitution in those with only a limited response to corticosteroids is a research arena that remains rife with opportunity and urgent clinical need.
Collapse
|