1
|
Gail LM, Schell KJ, Łacina P, Strobl J, Bolton SJ, Steinbakk Ulriksen E, Bogunia-Kubik K, Greinix H, Crossland RE, Inngjerdingen M, Stary G. Complex interactions of cellular players in chronic Graft-versus-Host Disease. Front Immunol 2023; 14:1199422. [PMID: 37435079 PMCID: PMC10332803 DOI: 10.3389/fimmu.2023.1199422] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/07/2023] [Indexed: 07/13/2023] Open
Abstract
Chronic Graft-versus-Host Disease is a life-threatening inflammatory condition that affects many patients after allogeneic hematopoietic stem cell transplantation. Although we have made substantial progress in understanding disease pathogenesis and the role of specific immune cell subsets, treatment options are still limited. To date, we lack a global understanding of the interplay between the different cellular players involved, in the affected tissues and at different stages of disease development and progression. In this review we summarize our current knowledge on pathogenic and protective mechanisms elicited by the major involved immune subsets, being T cells, B cells, NK cells and antigen presenting cells, as well as the microbiome, with a special focus on intercellular communication of these cell types via extracellular vesicles as up-and-coming fields in chronic Graft-versus-Host Disease research. Lastly, we discuss the importance of understanding systemic and local aberrant cell communication during disease for defining better biomarkers and therapeutic targets, eventually enabling the design of personalized treatment schemes.
Collapse
Affiliation(s)
- Laura Marie Gail
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kimberly Julia Schell
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Steven J. Bolton
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Hildegard Greinix
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Rachel Emily Crossland
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|
2
|
Khanolkar RA, Tripathi G, Dharmani-Khan P, Dabas R, Kinzel M, Kalra A, Puckrin R, Jimenez-Zepeda V, Jamani K, Duggan PR, Chaudhry A, Bryant A, Stewart DA, Khan FM, Storek J. Incomplete chimerism following myeloablative and anti-thymocyte globulin-conditioned hematopoietic cell transplantation is a risk factor for relapse and chronic graft-versus-host disease. Cytotherapy 2022; 24:1225-1231. [PMID: 36057497 DOI: 10.1016/j.jcyt.2022.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/16/2022] [Accepted: 07/31/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AIMS The value of routine chimerism determination after myeloablative hematopoietic cell transplantation (HCT) is unclear, particularly in the setting of anti-thymocyte globulin (ATG)-based graft-versus-host disease (GVHD) prophylaxis. METHODS Blood samples were collected at 3 months post-HCT from 558 patients who received myeloablative conditioning and ATG-based GVHD prophylaxis. Chimerism was assessed using multiplex polymerase chain reaction of short tandem repeats in sorted T cells (CD3+) and leukemia lineage cells (CD13+CD33+ for myeloid malignancies and CD19+ for B-lymphoid malignancies). ATG exposure was determined using a flow cytometry-based assay. The primary outcomes of interest were relapse and chronic GVHD (cGVHD). RESULTS Incomplete (<95%) T-cell chimerism and leukemia lineage chimerism were present in 17% and 4% of patients, respectively. Patients with incomplete T-cell chimerism had a significantly greater incidence of relapse (36% versus 22%, subhazard ratio [SHR] = 2.03, P = 0.001) and lower incidence of cGVHD (8% versus 25%, SHR = 0.29, P < 0.001) compared with patients with complete chimerism. In multivariate modeling, patients with high post-transplant ATG area under the curve and any cytomegalovirus (CMV) serostatus other than donor/recipient seropositivity (non-D+R+) had an increased likelihood of incomplete T-cell chimerism. Patients with incomplete leukemia lineage chimerism had a significantly greater incidence of relapse (50% versus 23%, SHR = 2.70, P = 0.011) and, surprisingly, a greater incidence of cGVHD (45% versus 20%, SHR = 2.64, P = 0.003). CONCLUSIONS High post-transplant ATG exposure and non-D+R+ CMV serostatus predispose patients to incomplete T-cell chimerism, which is associated with an increased risk of relapse. The increased risk of cGVHD with incomplete B-cell/myeloid chimerism is a novel finding that suggests an important role for recipient antigen-presenting cells in cGVHD pathogenesis.
Collapse
Affiliation(s)
- Rutvij A Khanolkar
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1.
| | - Gaurav Tripathi
- Department of Laboratory Medicine and Pathology, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Precision Laboratories, Calgary, Canada, T2N 4N1
| | - Poonam Dharmani-Khan
- Department of Laboratory Medicine and Pathology, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Precision Laboratories, Calgary, Canada, T2N 4N1
| | - Rosy Dabas
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1
| | - Megan Kinzel
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1
| | - Amit Kalra
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1
| | - Robert Puckrin
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Health Services, Calgary, Canada, T2N 4N1
| | - Victor Jimenez-Zepeda
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Health Services, Calgary, Canada, T2N 4N1
| | - Kareem Jamani
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Health Services, Calgary, Canada, T2N 4N1
| | - Peter R Duggan
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Health Services, Calgary, Canada, T2N 4N1
| | - Ahsan Chaudhry
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Health Services, Calgary, Canada, T2N 4N1
| | - Adam Bryant
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Health Services, Calgary, Canada, T2N 4N1
| | - Douglas A Stewart
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Health Services, Calgary, Canada, T2N 4N1
| | - Faisal M Khan
- Department of Laboratory Medicine and Pathology, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Precision Laboratories, Calgary, Canada, T2N 4N1
| | - Jan Storek
- Department of Medicine, University of Calgary, Calgary, Canada, T2N 4N1; Alberta Health Services, Calgary, Canada, T2N 4N1
| |
Collapse
|
3
|
Jardine L, Cytlak U, Gunawan M, Reynolds G, Green K, Wang XN, Pagan S, Paramitha M, Lamb CA, Long AK, Hurst E, Nair S, Jackson GH, Publicover A, Bigley V, Haniffa M, Simpson AJ, Collin M. Donor monocyte-derived macrophages promote human acute graft-versus-host disease. J Clin Invest 2021; 130:4574-4586. [PMID: 32453711 PMCID: PMC7456218 DOI: 10.1172/jci133909] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/19/2020] [Indexed: 12/16/2022] Open
Abstract
Myelopoiesis is invariably present and contributes to pathology in animal models of graft-versus-host disease (GVHD). In humans, a rich inflammatory infiltrate bearing macrophage markers has also been described in histological studies. In order to determine the origin, functional properties, and role in pathogenesis of these cells, we isolated single-cell suspensions from acute cutaneous GVHD and subjected them to genotype, transcriptome, and in vitro functional analysis. A donor-derived population of CD11c+CD14+ cells was the dominant population of all leukocytes in GVHD. Surface phenotype and NanoString gene expression profiling indicated the closest steady-state counterpart of these cells to be monocyte-derived macrophages. In GVHD, however, there was upregulation of monocyte antigens SIRPα and S100A8/9 transcripts associated with leukocyte trafficking, pattern recognition, antigen presentation, and costimulation. Isolated GVHD macrophages stimulated greater proliferation and activation of allogeneic T cells and secreted higher levels of inflammatory cytokines than their steady-state counterparts. In HLA-matched mixed leukocyte reactions, we also observed differentiation of activated macrophages with a similar phenotype. These exhibited cytopathicity to a keratinocyte cell line and mediated pathological damage to skin explants independently of T cells. Together, these results define the origin, functional properties, and potential pathogenic roles of human GVHD macrophages.
Collapse
Affiliation(s)
- Laura Jardine
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Urszula Cytlak
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Merry Gunawan
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Gary Reynolds
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Kile Green
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Sarah Pagan
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Maharani Paramitha
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher A Lamb
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Anna K Long
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Erin Hurst
- Northern Centre for Bone Marrow Transplantation and
| | - Smeera Nair
- Northern Centre for Bone Marrow Transplantation and
| | - Graham H Jackson
- Northern Centre for Bone Marrow Transplantation and.,Northern Institute of Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Amy Publicover
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Venetia Bigley
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Muzlifah Haniffa
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - A J Simpson
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Matthew Collin
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
4
|
Kubota N, Saito A, Tanaka R, Nakamura Y, Watanabe R, Fujisawa Y, Ishitsuka Y, Clausen BE, Fujimoto M, Okiyama N. Langerhans Cells Suppress CD8 + T Cells In Situ during Mucocutaneous Acute Graft-Versus-Host Disease. J Invest Dermatol 2020; 141:1177-1187.e3. [PMID: 33091425 DOI: 10.1016/j.jid.2020.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 08/29/2020] [Accepted: 09/02/2020] [Indexed: 02/02/2023]
Abstract
Acute graft-versus-host disease (aGVHD) induced by allogenic hematopoietic stem cell transplantation is an immunological disorder in which donor lymphocytes attack recipient organs. It has been proven that recipient nonhematopoietic tissue cells, such as keratinocytes, are sufficient as immunological targets for allogenic donor T cells, whereas Langerhans cells (LCs) are potent professional hematopoietic antigen-presenting cells existing in the target epidermis and eliminated during the early phase of mucocutaneous aGVHD. Moreover, LCs have been reported to negatively regulate various types of immune responses. Here, we present data showing that initial depletion of recipient LCs exacerbates mucocutaneous lesions in a murine model of allogenic bone marrow transplantation-induced aGVHD. Furthermore, another murine model of mucocutaneous aGVHD induced in mice with keratinocytes genetically expressing chicken ovalbumin by transfer of ovalbumin-specific CD8+ OT-I cells also showed that LC-depleted recipient mice develop aggravated mucocutaneous disease owing to decreased apoptosis of skin-infiltrating OT-I cells. Moreover, coexisting LCs directly induce apoptosis and inhibit the proliferation of OT-I cells in vitro partially via B7 family proteins. Collectively, our results indicate that LCs negatively regulate mucocutaneous aGVHD-like lesions in situ by inhibiting the number of infiltrating CD8+ T cells.
Collapse
Affiliation(s)
- Noriko Kubota
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akimasa Saito
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ryota Tanaka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshiyuki Nakamura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Rei Watanabe
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yosuke Ishitsuka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Naoko Okiyama
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
5
|
Tijaro-Ovalle NM, Karantanos T, Wang HT, Boussiotis VA. Metabolic Targets for Improvement of Allogeneic Hematopoietic Stem Cell Transplantation and Graft-vs.-Host Disease. Front Immunol 2019; 10:295. [PMID: 30891031 PMCID: PMC6411635 DOI: 10.3389/fimmu.2019.00295] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 02/05/2019] [Indexed: 12/13/2022] Open
Abstract
Utilization of the adaptive immune system against malignancies, both by immune-based therapies to activate T cells in vivo to attack cancer and by T-cell therapies to transfer effector cytolytic T lymphocytes (CTL) to the cancer patient, represent major novel therapeutic advancements in oncologic therapy. Allogeneic hematopoietic stem cell (HSC) transplantation (HSCT) is a form of cell-based therapy, which replaces the HSC in the patient's bone marrow but also serves as a T-cell therapy due to the Graft-vs.-leukemia (GVL) effect mediated by donor T cells transferred with the graft. Allogeneic HSCT provides one potentially curative option to patients with relapsed or refractory leukemia but Graft-vs.-Host-Disease (GVHD) is the main cause of non-relapse mortality and limits the therapeutic benefit of allogeneic HSCT. Metabolism is a common cellular feature and has a key role in the differentiation and function of T cells during the immune response. Naïve T cells and memory T cells that mediate GVHD and GVL, respectively, utilize distinct metabolic programs to obtain their immunological and functional specification. Thus, metabolic targets that mediate immunosuppression might differentially affect the functional program of GVHD-mediating or GVL-mediating T cells. Components of the innate immune system that are indispensable for the activation of alloreactive T cells are also subjected to metabolism-dependent regulation. Metabolic alterations have also been implicated in the resistance to chemotherapy and survival of malignant cells such as leukemia and lymphoma, which are targeted by GVL-mediating T cells. Development of novel approaches to inhibit the activation of GVHD-specific naïve T cell but maintain the function of GVL-specific memory T cells will have a major impact on the therapeutic benefit of HSCT. Here, we will highlight the importance of metabolism on the function of GVHD-inducing and GVL-inducing alloreactive T cells as well as on antigen presenting cells (APC), which are required for presentation of host antigens. We will also analyze the metabolic alterations involved in the leukemogenesis which could differentiate leukemia initiating cells from normal HSC, providing potential therapeutic opportunities. Finally, we will discuss the immuno-metabolic effects of key drugs that might be repurposed for metabolic management of GVHD without compromising GVL.
Collapse
Affiliation(s)
- Natalia M Tijaro-Ovalle
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Theodoros Karantanos
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Hong-Tao Wang
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
6
|
Radojcic V, Paz K, Chung J, Du J, Perkey ET, Flynn R, Ivcevic S, Zaiken M, Friedman A, Yan M, Pletneva MA, Sarantopoulos S, Siebel CW, Blazar BR, Maillard I. Notch signaling mediated by Delta-like ligands 1 and 4 controls the pathogenesis of chronic GVHD in mice. Blood 2018; 132:2188-2200. [PMID: 30181175 PMCID: PMC6238189 DOI: 10.1182/blood-2018-03-841155] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a major complication of allogeneic hematopoietic cell transplantation (allo-HCT) and remains an area of unmet clinical need with few treatment options available. Notch blockade prevents acute GVHD in multiple mouse models, but the impact of Notch signaling on cGVHD remains unknown. Using genetic and antibody-mediated strategies of Notch inhibition, we investigated the role of Notch signaling in complementary mouse cGVHD models that mimic several aspects of human cGVHD in search of candidate therapeutics. In the B10.D2→BALB/c model of sclerodermatous cGVHD, Delta-like ligand 4 (Dll4)-driven Notch signaling was essential for disease development. Antibody-mediated Dll4 inhibition conferred maximum benefits when pursued early in a preventative fashion, with anti-Dll1 enhancing early protection. Notch-deficient alloantigen-specific T cells showed no early defects in proliferation or helper polarization in vivo but subsequently exhibited markedly decreased cytokine secretion and enhanced accumulation of FoxP3+ regulatory T cells. In the B6→B10.BR major histocompatibility complex-mismatched model with multi-organ system cGVHD and prominent bronchiolitis obliterans (BO), but not skin manifestations, absence of Notch signaling in T cells provided long-lasting disease protection that was replicated by systemic targeting of Dll1, Dll4, or both Notch ligands, even during established disease. Notch inhibition decreased target organ damage and germinal center formation. Moreover, decreased BO-cGVHD was observed upon inactivation of Notch1 and/or Notch2 in T cells. Systemic targeting of Notch2 alone was safe and conferred therapeutic benefits. Altogether, Notch ligands and receptors regulate key pathogenic steps in cGVHD and emerge as novel druggable targets to prevent or treat different forms of cGVHD.
Collapse
Affiliation(s)
- Vedran Radojcic
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Katelyn Paz
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Jooho Chung
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
| | - Jing Du
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Eric T Perkey
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
| | - Ryan Flynn
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Sanja Ivcevic
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Michael Zaiken
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Ann Friedman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Minhong Yan
- Department of Discovery Oncology, Genentech, South San Francisco, CA
| | | | - Stefanie Sarantopoulos
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC; and
| | | | - Bruce R Blazar
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Ivan Maillard
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
7
|
Cancer Cell Death-Inducing Radiotherapy: Impact on Local Tumour Control, Tumour Cell Proliferation and Induction of Systemic Anti-tumour Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 930:151-72. [PMID: 27558821 DOI: 10.1007/978-3-319-39406-0_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Radiotherapy (RT) predominantly is aimed to induce DNA damage in tumour cells that results in reduction of their clonogenicity and finally in tumour cell death. Adaptation of RT with higher single doses has become necessary and led to a more detailed view on what kind of tumour cell death is induced and which immunological consequences result from it. RT is capable of rendering tumour cells immunogenic by modifying the tumour cell phenotype and the microenvironment. Danger signals are released as well as the senescence-associated secretory phenotype. This results in maturation of dendritic cells and priming of cytotoxic T cells as well as in activation of natural killer cells. However, RT on the other hand can also result in immune suppressive events including apoptosis induction and foster tumour cell proliferation. That's why RT is nowadays increasingly combined with selected immunotherapies.
Collapse
|
8
|
Toubai T, Mathewson ND, Magenau J, Reddy P. Danger Signals and Graft-versus-host Disease: Current Understanding and Future Perspectives. Front Immunol 2016; 7:539. [PMID: 27965667 PMCID: PMC5126092 DOI: 10.3389/fimmu.2016.00539] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 11/15/2016] [Indexed: 12/22/2022] Open
Abstract
Graft-versus-host response after allogeneic hematopoietic stem cell transplantation (allo-HCT) represents one of the most intense inflammatory responses observed in humans. Host conditioning facilitates engraftment of donor cells, but the tissue injury caused from it primes the critical first steps in the development of acute graft-versus-host disease (GVHD). Tissue injuries release pro-inflammatory cytokines (such as TNF-α, IL-1β, and IL-6) through widespread stimulation of pattern recognition receptors (PRRs) by the release of danger stimuli, such as damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs). DAMPs and PAMPs function as potent stimulators for host and donor-derived antigen presenting cells (APCs) that in turn activate and amplify the responses of alloreactive donor T cells. Emerging data also point towards a role for suppression of DAMP induced inflammation by the APCs and donor T cells in mitigating GVHD severity. In this review, we summarize the current understanding on the role of danger stimuli, such as the DAMPs and PAMPs, in GVHD.
Collapse
Affiliation(s)
- Tomomi Toubai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| | - Nathan D Mathewson
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute , Boston, MA , USA
| | - John Magenau
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| | - Pavan Reddy
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| |
Collapse
|
9
|
Collin M, Jardine L. A question of persistence: Langerhans cells and graft-versus-host disease. Exp Dermatol 2015; 23:234-5. [PMID: 24443966 PMCID: PMC4150527 DOI: 10.1111/exd.12325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2014] [Indexed: 01/03/2023]
Abstract
Langerhans cells (LCs) have been scrutinized many times in studies of the pathogenesis of graft-versus-host disease (GVHD). As migratory dendritic cells, LCs are capable of direct antigen presentation to cytotoxic T cells. Their self-renewal capacity has led to speculation that persistent recipient LCs could provide a continuous source of host antigen to donor T cells infused during hematopoietic stem cell transplantation (HSCT). In this issue of Experimental Dermatology, a new study examines at the relationship between recipient LCs and chronic GVHD.
Collapse
Affiliation(s)
- Matthew Collin
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | |
Collapse
|
10
|
Galvani RG, Lemos R, Areal RB, Salvador PA, Zamboni DS, Wanderley JLM, Bonomo A. Disease severity and mortality can be independently regulated in a mouse model of experimental graft versus host disease. PLoS One 2015; 10:e0118079. [PMID: 25643148 PMCID: PMC4313938 DOI: 10.1371/journal.pone.0118079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 01/05/2015] [Indexed: 12/16/2022] Open
Abstract
Graft versus host disease (GVHD) is the major limitation of allogeneic hematopoietic stem cell transplantation (HSCT) presenting high mortality and morbidity rates. However, the exact cause of death is not completely understood and does not correlate with specific clinical and histological parameters of disease. Here we show, by using a semi-allogeneic mouse model of GVHD, that mortality and morbidity can be experimentally separated. We injected bone marrow-derived dendritic cells (BMDC) from NOD2/CARD15-deficient donors into semi-allogeneic irradiated chimaeras and observed that recipients were protected from death. However, no protection was observed regarding clinical or pathological scores up to 20 days after transplantation. Protection from death was associated with decreased bacterial translocation, faster hematologic recovery and epithelial integrity maintenance despite mononuclear infiltration at day 20 post-GVHD induction with no skew towards different T helper phenotypes. The protected mice recovered from aGVHD and progressively reached scores compatible with healthy animals. Altogether, our data indicate that severity and mortality can be separate events providing a model to study transplant-related mortality.
Collapse
Affiliation(s)
- Rômulo G. Galvani
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ramon Lemos
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rômulo B. Areal
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Pollyanna A. Salvador
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Dario S. Zamboni
- Departamento de Biologia Celular, Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - João Luiz M. Wanderley
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- NUPEM, Campus Macaé Professor Aloísio Teixeira, Universidade Federal do Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
- * E-mail:
| | - Adriana Bonomo
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratorio de Pesquisa sobre o Timo, Instituo Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Langerhans cell homeostasis and turnover after nonmyeloablative and myeloablative allogeneic hematopoietic cell transplantation. Transplantation 2014; 98:563-8. [PMID: 24717220 DOI: 10.1097/tp.0000000000000097] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Langerhans cells (LCs) are self-renewing epidermal myeloid cells that can migrate and mature into dendritic cells. Recipient LCs that survive cytotoxic therapy given in preparation for allogeneic hematopoietic cell transplantation may prime donor T cells to mediate cutaneous graft-versus-host disease (GVHD). This possible association, however, has not been investigated in the setting of nonmyeloablative allografting. METHODS We prospectively studied the kinetics of LC-chimerism after sex-mismatched allogeneic hematopoietic cell transplantation with nonmyeloablative (n=23) or myeloablative (n=25) conditioning. Combined XY-FISH and Langerin-staining was used to assess donor LC-chimerism in skin biopsies obtained on days 28, 56, and 84 after transplant. The degree of donor LC-chimerism was correlated with the development of skin GVHD. RESULTS We observed significantly delayed donor LC-engraftment after nonmyeloablative transplantation compared with other hematopoietic compartments and compared with LC-engraftment after myeloablative conditioning. In most recipients of nonmyeloablative transplants, recipient LCs proliferated in situ, recruitment of donor-LCs was delayed by two months, and full donor LC-chimerism was only reached by day 84 after transplant. Although persistence of host LCs on day-28 after transplant was not predictive for acute or chronic skin GVHD, the recruitment of donor-derived LCs was associated with nonspecific inflammatory infiltrates (P=0.009). CONCLUSIONS These results show that LCs can self-renew locally but are replaced by circulating precursors even after minimally toxic nonmyeloablative transplant conditioning. Cutaneous inflammation accompanies donor LC-engraftment, but differences in LC conversion-kinetics do not predict clinical or histopathological GVHD.
Collapse
|
12
|
Andani R, Robertson I, Macdonald KPA, Durrant S, Hill GR, Khosrotehrani K. Origin of Langerhans cells in normal skin and chronic GVHD after hematopoietic stem-cell transplantation. Exp Dermatol 2014; 23:75-7. [PMID: 24313654 DOI: 10.1111/exd.12301] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2013] [Indexed: 12/28/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is a common complication following allogeneic stem-cell transplantation (SCT). Past studies have implicated the persistence of host antigen-presenting cells (APCs) in GVHD. Our objective was to determine the frequency of host Langerhans cells (LCs) in normal skin post-SCT and ask if their persistence could predict cGVHD. Biopsies of normal skin from 124 sex-mismatched T-cell-replete allogenic SCT recipients were taken 100 days post-transplant. Patients with acute GVHD and those with <9 months of follow-up were excluded and prospective follow-up information was collected from remaining 22 patients. CD1a staining and X and Y chromosome in-situ hybridization were performed to label LCs and to identify their host or donor origin. At 3 months, 59 ± 5% of LCs were host derived. The density of LCs and the proportion of host-derived LCs were similar between patients that did or did not develop cGVHD. Most LCs in the skin remained of host origin 3 months after SCT regardless of cGVHD status. This finding is in line with the redundant role of LCs in acute GVHD initiation uncovered in recent experimental models.
Collapse
Affiliation(s)
- Rafiq Andani
- Experimental Dermatology Laboratory, UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | | | | | | | | | | |
Collapse
|
13
|
Waller EK, Logan BR, Harris WAC, Devine SM, Porter DL, Mineishi S, McCarty JM, Gonzalez CE, Spitzer TR, Krijanovski OI, Linenberger ML, Woolfrey A, Howard A, Wu J, Confer DL, Anasetti C. Improved survival after transplantation of more donor plasmacytoid dendritic or naïve T cells from unrelated-donor marrow grafts: results from BMTCTN 0201. J Clin Oncol 2014; 32:2365-72. [PMID: 24982459 DOI: 10.1200/jco.2013.54.4577] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To characterize relationships between specific immune cell subsets in bone marrow (BM) or granulocyte colony-stimulating factor-mobilized peripheral blood (PB) stem cells collected from unrelated donors and clinical outcomes of patients undergoing transplantation in BMTCTN 0201. PATIENTS AND METHODS Fresh aliquots of 161 BM and 147 PB stem-cell allografts from North American donors randomly assigned to donate BM or PB stem cells and numbers of transplanted cells were correlated with overall survival (OS), relapse, and graft-versus-host disease (GvHD). RESULTS Patients with evaluable grafts were similar to all BMTCTN 0201 patients. The numbers of plasmacytoid dendritic cells (pDCs) and naïve T cells (Tns) in BM allografts were independently associated with OS in multivariable analyses including recipient and donor characteristics, such as human leukocyte antigen mismatch, age, and use of antithymocyte globulin. BM recipients of > median number of pDCs, naïve CD8(+) T cells (CD8Tns), or naïve CD4(+) T cells (CD4Tns) had better 3-year OS (pDCs, 56% v 35%; P = .025; CD8Tns, 56% v 37%; P = .012; CD4Tns, 55% v 37%; P = .009). Transplantation of more BM Tns was associated with less grade 3 to 4 acute GvHD but similar rates of relapse. Transplantation of more BM pDCs was associated with fewer deaths resulting from GvHD or from graft rejection. Analysis of PB grafts did not identify a donor cell subset significantly associated with OS, relapse, or GvHD. CONCLUSION Donor immune cells in BM but not PB stem-cell grafts were associated with survival after unrelated-donor allogeneic hematopoietic stem-cell transplantation. The biologic activity of donor immune cells in allogeneic transplantation varied between graft sources. Donor grafts with more BM-derived Tns and pDCs favorably regulated post-transplantation immunity in allogeneic hematopoietic stem-cell transplantation.
Collapse
Affiliation(s)
- Edmund K Waller
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL.
| | - Brent R Logan
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Wayne A C Harris
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Steven M Devine
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - David L Porter
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Shin Mineishi
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - John M McCarty
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Corina E Gonzalez
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Thomas R Spitzer
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Oleg I Krijanovski
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Michael L Linenberger
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Ann Woolfrey
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Alan Howard
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Juan Wu
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Dennis L Confer
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Claudio Anasetti
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
14
|
Oviedo A, Yañez R, Colmenero I, Aldea M, Rubio A, Bueren JA, Lamana ML. Reduced efficacy of mesenchymal stromal cells in preventing graft-versus-host disease in an in vivo model of haploidentical bone marrow transplant with leukemia. Cell Transplant 2012; 22:1381-94. [PMID: 23044223 DOI: 10.3727/096368912x657666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cell (MSC) immunosuppressive properties have been applied to treat graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplants (HSCTs). We have previously demonstrated that MSC infusions early after haplo-HSCT prevent GVHD in a haploidentical-HSCT mouse model. Now, we investigated the impact that MSCs' immunosuppressive properties have on the graft-versus-leukemia (GVL) effect. First, to mimic a chronic myeloid leukemia (CML) relapse after a haploidentical HSCT, lethally irradiated mice were coinfused with haploidentical donor bone marrow cells plus syngenic hematopoietic progenitors transduced with a retroviral vector encoding both the BCR/ABL oncogene and the ΔNGFR marker gene. As expected, a CML-like myeloproliferative syndrome developed in all the recipient animals. The addition of haploidentical splenocytes to the transplanted graft prevented CML development by a GVL effect, and all transplanted recipients died of GVHD. This GVL mouse model allowed us to investigate the impact of MSCs infused to prevent GVHD on days 0, 7, and 14 after HSCT, on the GVL effect, expecting an increase in leukemic relapse. Strikingly, a high mortality of the recipients was observed, caused by GVHD, and only few leukemic cells were detected in the recipient animals. In contrast, GVHD prevention by MSCs in the absence of BCR/ABL leukemic cells resulted in a significant survival of the recipients. In vitro data pointed to an inability of MSCs to control strong CTLs responses against BCR/ABL. Our results show that, although an evident increase in leukemic relapses induced by MSCs could not be detected, they showed a reduced efficacy in preventing GVHD that precluded us to draw clear conclusions on MSCs' impact over GVL effect.
Collapse
Affiliation(s)
- Alberto Oviedo
- Hematopoiesis and Gene Therapy Division, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
15
|
Dendritic cells and regulation of graft-versus-host disease and graft-versus-leukemia activity. Blood 2012; 119:5088-103. [PMID: 22403259 DOI: 10.1182/blood-2011-11-364091] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hematopoietic stem cell transplantation is the only curative treatment for many malignant hematologic diseases, with an often critical graft-versus-leukemia effect. Despite peritransplant prophylaxis, GVHD remains a significant cause of posthematopoietic stem cell transplantation morbidity and mortality. Traditional therapies have targeted T cells, yet immunostimulatory dendritic cells (DCs) are critical in the pathogenesis of GVHD. Furthermore, DCs also have tolerogenic properties. Monitoring of DC characteristics may be predictive of outcome, and therapies that target DCs are innovative and promising. DCs may be targeted in vivo or tolerogenic (tol) DCs may be generated in vitro and given in the peritransplant period. Other cellular therapies, notably regulatory T cells (T(reg)) and mesenchymal stem cells, mediate important effects through DCs and show promise for the prevention and treatment of GVHD in early human studies. Therapies are likely to be more effective if they have synergistic effects or target both DCs and T cells in vivo, such as tolDCs or T(reg). Given the effectiveness of tolDCs in experimental models of GVHD and their safety in early human studies for type 1 diabetes, it is crucial that tolDCs be investigated in the prevention and treatment of human GVHD while ensuring conservation of graft-versus-leukemia effects.
Collapse
|
16
|
Castor MGM, Pinho V, Teixeira MM. The role of chemokines in mediating graft versus host disease: opportunities for novel therapeutics. Front Pharmacol 2012; 3:23. [PMID: 22375119 PMCID: PMC3285883 DOI: 10.3389/fphar.2012.00023] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 02/08/2012] [Indexed: 11/25/2022] Open
Abstract
Bone marrow transplantation (BMT) is the current therapy of choice for several malignancies and severe autoimmune diseases. Graft versus host disease (GVHD) is the major complication associated with BMT. T lymphocytes and other leukocytes migrate into target organs during GVHD, become activated and mediate tissue damage. Chemokines are well known inducers of leukocyte trafficking and activation and contribute to the pathogenesis of GVHD. Here, we review the major animal models used to study GVHD and the role of chemokines in mediating tissue damage in these models. The role of these molecules in promoting potential beneficial effects of the graft, especially graft versus leukemia, is also discussed. Finally, the various pharmacological strategies to block the chemokine system or downstream signaling events in the context of GVHD are discussed.
Collapse
Affiliation(s)
- Marina G M Castor
- Immunopharmacology, Department of Immunology and Biochemistry, Institute of Biological Sciences, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | | | | |
Collapse
|
17
|
Miao X, Luo D, Min W, Lin X, Wang D, Xu Y, Wu D. Potential efficacy of imiquimod on immunity-related cytokines in murine skin in vivo and in human Langerhans cells in vitro. Int J Dermatol 2012; 51:1116-22. [PMID: 22348684 DOI: 10.1111/j.1365-4632.2011.05382.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Imiquimod, an immune-response modifier, has been proven to be clinically effective in the treatment of viral infections and skin cancers, but its mechanism of action remains poorly understood. This study aimed to assess the effects of imiquimod on the expression of three immunity-related cytokines, TNF-α, IL-1β, and IL-6. MATERIALS AND METHODS Female BALB/C mice were treated for seven days with topical 1% imiquimod cream; they were then killed and skin samples were snap-frozen. In the in vitro studies, both purified LCs and HaCaT cells were incubated with 5 μg/ml imiquimod for four hours. In all samples, the mRNA levels of TNF-α, IL-1β, and IL-6 were then detected by reverse transcription polymerase chain reaction, and the secretion levels were determined by an enzyme-linked immunosorbent assay. RESULTS Imiquimod upregulated the mRNA and protein expression levels of TNF-α, IL-1β, and IL-6 in the skin of imiquimod-treated BALB/C mice and in human LCs, compared with untreated controls (P<0.05). However, there was no significant difference in the expression of these cytokines in imiquimod-treated and untreated HaCaT cells. CONCLUSIONS These findings indicate that imiquimod increased the expression of TNF-α, IL-1β, and IL-6 in skin and that the target cell of imiquimod may be the LCs but is unlikely to be the epidermal keratinocytes.
Collapse
Affiliation(s)
- Xu Miao
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
18
|
The humanized anti-HLA-DR moAb, IMMU-114, depletes APCs and reduces alloreactive T cells: implications for preventing GVHD. Bone Marrow Transplant 2011; 47:967-80. [PMID: 22020022 DOI: 10.1038/bmt.2011.203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In contrast to the conventional immunosuppressive agents and nonselective T-cell-depleting antibodies, selective depletion of donor alloreactive T cells and/or host APCs, particularly DCs, represents a novel approach that can effectively control GVHD with less or no impairment of T-cell-mediated antiviral and GVL immunity. Here we report that IMMU-114, a humanized anti-human leukocyte antigen-DR (HLA-DR) moAb, efficiently depleted human PBMCs of all APCs, including B cells, monocytes, myeloid DC type-1 (mDC1), mDC2 and plasmacytoid DCs (pDCs). Early and late apoptosis of mDC1, mDC2 and pDCs, and late apoptosis of all APC subsets, were increased by IMMU-114 treatment. Although IMMU-114 had little, if any, effect on the survival and apoptosis of non-B lymphocytes (>80% of which are T cells and ∼1-2% of T cells express HLA-DR), it selectively inhibited the proliferation of purified HLA-DR(+) T cells rather than HLA-DR(-) T cells. As a consequence, IMMU-114 treatment resulted in suppressed T-cell proliferation and reduced CD25(+) alloreactive T cells in allogeneic MLRs. Given the critical roles of APCs and alloreactive T cells in the pathogenesis of GVHD, these results suggest that IMMU-114 may have therapeutic potential against GVHD.
Collapse
|
19
|
Whole-body UVB irradiation during allogeneic hematopoietic cell transplantation is safe and decreases acute graft-versus-host disease. J Invest Dermatol 2011; 132:179-87. [PMID: 21850024 DOI: 10.1038/jid.2011.255] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Depletion of host Langerhans cells (LCs) prevents cutaneous graft-versus-host disease (GvHD) in mice. We analyzed whether UVB irradiation is tolerated during the course of human allogeneic hematopoietic cell transplantation and whether depletion of LCs by broadband UVB could improve GvHD outcome. A total of 17 patients received six whole-body UVB irradiations with 75% of the individually determined minimal erythemal dose after conditioning with a reduced intensity protocol. LCs, dermal dendritic cells (DCs), and macrophages were analyzed before and after UVB irradiation by immunohistochemical analysis. Circulating blood cells and serum factors were analyzed in parallel. In striking contrast to previous data, our irradiation protocol was well tolerated in all patients. UVB treatment decreased the number of LCs and also affected dermal DCs. UVB-treated patients also had significantly higher 25-hydroxyvitamin D3 serum levels and higher numbers of circulating CD4+ FoxP3+ regulatory T cells. Strikingly, nine out of nine patients with complete LC depletion (<1 LC per field) developed only grade I GvHD or no GvHD up to day 100. Our results strongly suggest that prophylactic UVB irradiation post transplant is safe and should be further explored as a clinical strategy to prevent acute (skin) GvHD.
Collapse
|
20
|
Hemmerling J, Wegner-Kops J, von Stebut E, Wolff D, Wagner EM, Hartwig UF, André MC, Theobald M, Schopf RE, Herr W, Meyer RG. Human epidermal Langerhans cells replenish skin xenografts and are depleted by alloreactive T cells in vivo. THE JOURNAL OF IMMUNOLOGY 2011; 187:1142-9. [PMID: 21697461 DOI: 10.4049/jimmunol.1001491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epidermal Langerhans cells (LC) are potent APCs surveying the skin. They are crucial regulators of T cell activation in the context of inflammatory skin disease and graft-versus-host disease (GVHD). In contrast to other dendritic cell subtypes, murine LC are able to reconstitute after local depletion without the need of peripheral blood-derived precursors. In this study, we introduce an experimental model of human skin grafted to NOD-SCID IL2Rγ(null) mice. In this model, we demonstrate that xenografting leads to the transient loss of LC from the human skin grafts. Despite the lack of a human hematopoietic system, human LC repopulated the xenografts 6 to 9 wk after transplantation. By staining of LC with the proliferation marker Ki67, we show that one third of the replenishing LC exhibit proliferative activity in vivo. We further used the skin xenograft as an in vivo model for human GVHD. HLA-disparate third-party T cells stimulated with skin donor-derived dendritic cells were injected intravenously into NOD-SCID IL2Rγ(null) mice that had been transplanted with human skin. The application of alloreactive T cells led to erythema and was associated with histological signs of GVHD limited to the transplanted human skin. The inflammation also led to the depletion of LC from the epidermis. In summary, we provide evidence that human LC are able to repopulate the skin independent of blood-derived precursor cells and that this at least partly relates to their proliferative capacity. Our data also propose xeno-transplantation of human skin as a model system for studying the role of skin dendritic cells in the efferent arm of GVHD.
Collapse
Affiliation(s)
- Julia Hemmerling
- Department of Hematology, Oncology, and Pneumology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Sclerodermatous chronic graft‐versus‐host disease induced by host T‐cell‐mediated autoimmunity. Immunol Cell Biol 2011; 90:358-67. [DOI: 10.1038/icb.2011.46] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
Harnessing dendritic cells to improve allogeneic hematopoietic cell transplantation outcome. Semin Immunol 2011; 23:50-7. [DOI: 10.1016/j.smim.2011.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 01/05/2011] [Indexed: 11/20/2022]
|
23
|
Abstract
Graft-versus-host disease (GVHD) is initiated and maintained by antigen-presenting cells (APCs) that prime alloreactive donor T cells. APCs are therefore attractive targets for GVHD prevention and treatment. APCs are diverse in phenotype and function, making understanding how APC subsets contribute to GVHD necessary for the development of APC-targeted therapies. Langerhans cells (LCs) have been shown to be sufficient to initiate skin GVHD in a major histocompatibility complex-mismatched model; however, their role when other host APC subsets are intact is unknown. To address this question, we used mice genetically engineered to be deficient in LCs by virtue of expression of diphtheria toxin A under the control of a BAC (bacterial artificial chromosome) transgenic hu-man Langerin locus. Neither CD8- nor CD4-mediated GVHD was diminished in recipients lacking LCs. Similarly, CD8- and CD4-mediated GVHD, including that in the skin, was unaffected if bone marrow came from donors that could not generate LCs, even though donor LCs engrafted in control mice. Engraftment of donor LCs after irradiation in wild-type hosts required donor T cells, with immunofluorescence revealing patches of donor and residual host LCs. Surprisingly, donor LC engraftment in Langerin-diphtheria toxin A (DTA) transgenic hosts was independent of donor T cells, suggesting that a Langerin(+) cell regulates repopulation of the LC compartment.
Collapse
|
24
|
Alyea EP, DeAngelo DJ, Moldrem J, Pagel JM, Przepiorka D, Sadelin M, Young JW, Giralt S, Bishop M, Riddell S. NCI First International Workshop on The Biology, Prevention and Treatment of Relapse after Allogeneic Hematopoietic Cell Transplantation: report from the committee on prevention of relapse following allogeneic cell transplantation for hematologic malignancies. Biol Blood Marrow Transplant 2010; 16:1037-69. [PMID: 20580849 PMCID: PMC3235046 DOI: 10.1016/j.bbmt.2010.05.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Accepted: 05/14/2010] [Indexed: 10/19/2022]
Abstract
Prevention of relapse after allogeneic hematopoietic stem cell transplantation is the most likely approach to improve survival of patients treated for hematologic malignancies. Herein we review the limits of currently available transplant therapies and the innovative strategies being developed to overcome resistance to therapy or to fill therapeutic modalities not currently available. These novel strategies include nonimmunologic therapies, such as targeted preparative regimens and posttransplant drug therapy, as well as immunologic interventions, including graft engineering, donor lymphocyte infusions, T cell engineering, vaccination, and dendritic cell-based approaches. Several aspects of the biology of the malignant cells as well as the host have been identified that obviate success of even these newer strategies. To maximize the potential for success, we recommend pursuing research to develop additional targeted therapies to be used in the preparative regimen or as maintenance posttransplant, better characterize the T cell and dendritic cells subsets involved in graft-versus-host disease and the graft-versus-leukemia/tumor effect, identify strategies for timing immunologic or nonimmunologic therapies to eliminate the noncycling cancer stem cell, identify more targets for immunotherapies, develop new vaccines that will not be limited by HLA, and develop methods to identify populations at very high risk for relapse to accelerate clinical development and avoid toxicity in patients not at risk for relapse.
Collapse
Affiliation(s)
- Edwin P Alyea
- Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Li JM, Southerland LT, Lu Y, Darlak KA, Giver CR, McMillin DW, Harris WAC, Jaye DL, Waller EK. Activation, immune polarization, and graft-versus-leukemia activity of donor T cells are regulated by specific subsets of donor bone marrow antigen-presenting cells in allogeneic hemopoietic stem cell transplantation. THE JOURNAL OF IMMUNOLOGY 2010; 183:7799-809. [PMID: 19933853 DOI: 10.4049/jimmunol.0900155] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We investigated the roles of specific subsets of donor APCs purified from bone marrow in donor T cell activation and graft-vs-leukemia (GvL) activity in murine models of hemopoietic stem cell transplantation. Lineage(-)CD11c(+) APC precursors were separated from donor bone marrow based on expression of CD11b. Transplanting lineage(-)CD11c(+)CD11b(-) APC (CD11b(-) APC) in combination with c-kit(+)Sca-1(+)lineage(-) hemopoietic stem cells (HSC) and congenic donor T cells led to increased donor CD4(+) and CD8(+) T cell proliferation and higher donor T cell chimerism than with transplanting grafts containing HSC, T cells, and lineage(-)CD11c(+)CD11b(+) APCs (CD11b(+) APC), or grafts containing only HSC and T cells. Transplanting CD11b(-) APCs induced Th1/type 1 cytotoxic T lymphocyte donor T cell immune polarization and enhanced GvL activity of donor T cells without increased graft-vs-host disease in both MHC- and minor histocompatibility Ag-mismatched murine hemopoietic stem cell transplantation models, whereas CD11b(+) APCs led to Th2/type 2 cytotoxic T lymphocyte donor T cell immune polarization. Donor CD11b(-) APCs were plasmacytoid dendritic cell progenitors (>90% CD317; PDCA-1(+)) and up-regulated CD80, CD86, and IL-12 during alloantigen presentation, whereas CD11b(+) APCs expressed Gr-1 and up-regulated expression of programmed death ligands-1 and 2 after activation. These results are the first to show that manipulation of the content of donor APCs in allogeneic HSC grafts can regulate donor T cell immunity and enhance GvL without increasing graft-vs-host disease activity.
Collapse
Affiliation(s)
- Jian-Ming Li
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
D'Orsogna LJ, Wright MP, Krueger RG, McKinnon EJ, Buffery SI, Witt CS, Staples N, Loh R, Cannell PK, Christiansen FT, French MA. Allogeneic hematopoietic stem cell transplantation recipients have defects of both switched and igm memory B cells. Biol Blood Marrow Transplant 2009; 15:795-803. [PMID: 19539210 DOI: 10.1016/j.bbmt.2008.11.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Accepted: 02/05/2009] [Indexed: 12/13/2022]
Abstract
Allogeneic hematopoietic stem cell transplant (HSCT) recipients were assessed to elucidate memory B cell defects underlying their increased susceptibility to infections, particularly by encapsulated bacteria. Circulating IgM memory B cells (CD19+, CD27+, IgM+) and switched memory B cells (CD19+, CD27+, IgM(-)) were enumerated in allogeneic HSCT recipients (n = 37) and healthy controls (n = 35). T lymphocyte subpopulations and serum levels of immunoglobulins, including IgG subclasses, and antibodies to pneumococcal polysaccharides were also assayed. Allogeneic HSCT recipients were deficient in both switched memory and IgM memory B cells compared to healthy controls (both P < .0001), irrespective of time post-HSCT. Switched memory B cell deficiency correlated with CD4+ T cell deficiency, and both correlated with serum levels of IgG1 (P < .0001), possibly reflecting impaired B cell isotype switching in germinal centres. "Steady-state" serum levels of antibodies to pneumococcal polysaccharides did not correlate with circulating memory B cells. Graft-versus-host disease (GVHD) was associated with lower IgM memory B cell counts and lower serum levels of IgG2, IgG4, IgA, and pneumococcal antibodies. The increased susceptibility of allogeneic HSCT patients to infection may reflect a combination of memory B cell defects, which are most common in patients with a history of GVHD.
Collapse
Affiliation(s)
- Lloyd J D'Orsogna
- Department of Clinical Immunology and Immunogenetics, Royal Perth Hospital and PathWest Laboratory Medicine, Perth, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhu KJ, Cen JP, Lou JX, Wang Q, Zhang X, Xu Y, Chen XZ, Cheng H. Imiquimod inhibits the differentiation but enhances the maturation of human monocyte-derived dendritic cells. Int Immunopharmacol 2009; 9:412-7. [PMID: 19185063 DOI: 10.1016/j.intimp.2008.12.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2008] [Revised: 12/09/2008] [Accepted: 12/29/2008] [Indexed: 10/21/2022]
Abstract
Imiquimod is a topically used immune response modifier effective in the treatment of genital warts caused by HPV. Its therapeutic effects are believed to be the release of proinflammatory cytokines from the monocyte-macrophage lineage resulting in a cascade of events abating the HPV replication. Dendritic cell maturation and activation have also been found to be induced by imiquimod. We hypothesized that imiquimod may promote the development of DC at all levels of their life cycle. In this study, in vitro cultured monocyte-derived dendritic cells (MoDC) were used to evaluate the effect of imiquimod regarding the modulation of DC differentiation, terminal maturation and their function by phenotypic cell surface molecules expression, cytokine secretion and T cell stimulation in allogeneic system. We demonstrate that imiquimod exerts differential effects on DC biology at different stages of DC development. It inhibits the differentiation of DC, which may indicate a more potent antigen uptake activity. DC maturation is induced by imiquimod with an enhanced antigen presenting activity and IL-12 production. These evidence might be relevant with the clinically proven effectiveness of imiquimod in the treatment of genital warts.
Collapse
Affiliation(s)
- Ke-Jian Zhu
- Department of Dermatology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW The aim of this article is to highlight both experimental and clinical studies contributing to our present understanding of acute graft versus host disease. RECENT FINDINGS New cellular players and new molecules in induction, modulation and treatment of graft versus host disease are reported. The contribution of antigen presenting cells to induction of graft versus host disease and graft versus leukemia effects is well understood in animal models and now needs confirmation in human analyses. Both, regulatory T cells and mesenchymal stem cells are a new hope for prophylaxis and treatment. Receptors of innate immunity, homing molecules and chemokines regulate activation and trafficking of these cellular players and may be used for selective blockade, activation or cell selection. SUMMARY Better understanding of the early events involved in graft versus host disease should allow risk adapted and more precise early or even preemptive treatment of graft versus host disease.
Collapse
Affiliation(s)
- Ernst Holler
- Department of Haematology/Oncology, University Medical Centre, Regensburg, Germany.
| |
Collapse
|
29
|
Young JW, Merad M, Hart DNJ. Dendritic cells in transplantation and immune-based therapies. Biol Blood Marrow Transplant 2007; 13:23-32. [PMID: 17222766 DOI: 10.1016/j.bbmt.2006.10.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Dendritic cells (DCs) are specialized, bone marrow-derived leukocytes critical to the onset of both innate and adaptive immunity. The divisions of labor among distinct human DC subtypes achieve the most effective balance between steady-state tolerance and the induction of innate and adaptive immunity against pathogens, tumors, and other insults. Maintenance of tolerance in the steady state is an active process involving resting or semimature DCs. Breakdowns in this homeostasis can result in autoimmunity. Perturbation of the steady state should first lead to the onset of innate immunity mediated by rapid responders in the form of plasmacytoid and monocyte-derived DC stimulators and natural killer (NK) and NK T-cell responders. These innate effectors then provide additional inflammatory cytokines, including interferon-gamma, which support the activation and maturation of resident and circulating populations of DCs. These are critical to the onset and expansion of adaptive immunity, including Th1, Th2, and cytotoxic T-lymphocyte responses. Rodent models are now revealing important data about distinct DC precursors, homeostasis of tissue-resident DCs, and DC turnover in response to inflammation and pathological conditions like graft-versus-host disease. The use of defined DC subtypes to stimulate both innate and adaptive immunity, either in combination or in a prime-boost vaccine sequence, may prove most useful clinically by harnessing both effector cell compartments.
Collapse
Affiliation(s)
- James W Young
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | | | |
Collapse
|
30
|
Durakovic N, Radojcic V, Powell J, Luznik L. Rapamycin Promotes Emergence of IL-10-Secreting Donor Lymphocyte Infusion-Derived T Cells Without Compromising Their Graft-Versus-Leukemia Reactivity. Transplantation 2007; 83:631-40. [PMID: 17353785 DOI: 10.1097/01.tp.0000256158.84418.ff] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There are limited data examining the effects of pharmacological immunosuppression on the in vivo fate of donor lymphocyte infusions (DLI)-derived T cells, their function, and their antitumor efficacy. METHODS We addressed this question in a murine model in which DLI is given to stable mixed chimeras resulting in lymphohematopoietic graft-versus-host (LH-GVH) response. In this model, LH-GVH potency can be directly measured as the kinetics of conversion to full donor chimerism and can be correlated with associated graft-versus-leukemia (GVL) reactivity. RESULTS We found discordance in DLI-mediated LH-GVH reactivity depending on the timing of rapamycin (RAPA) administration. Delayed administration of RAPA in contrast to its early administration at the time of adoptive transfer did not interfere with conversion to full donor chimerism. Moreover, delayed administration of RAPA preserved the GVL reactivity of DLI. Analysis of the long-term chimeras showed that regardless of RAPA administration, adoptively transferred T cells mediating the LH-GVH response contribute minimally to the reconstitution of the peripheral T-cell compartment and exhibit profound hyporesponsiveness and decreased production of interleukin (IL)-2 on restimulation in vitro. However, we observed only in the RAPA-treated chimeras that the remaining hyporesponsive DLI-derived CD4+ T cells secrete large amounts of IL-10, a known immunoregulatory cytokine. CONCLUSIONS We conclude that delayed administration of RAPA after DLI does not interfere with their LH-GVH reactivity but promotes the emergence of IL-10-secreting DLI-derived CD4+ T cells that might contribute to the drug's known ability to promote bilateral donor host tolerance without interfering with GVL reactivity.
Collapse
Affiliation(s)
- Nadira Durakovic
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | | | | |
Collapse
|
31
|
Chakraverty R, Sykes M. The role of antigen-presenting cells in triggering graft-versus-host disease and graft-versus-leukemia. Blood 2007; 110:9-17. [PMID: 17327406 PMCID: PMC1896131 DOI: 10.1182/blood-2006-12-022038] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
After allogeneic blood or bone marrow transplantation, donor T cells interact with a distorted antigen-presenting cell (APC) environment in which some, but not all, host APCs are replaced by APCs from the donor. Significantly, host APCs are required for the priming of acute graft-versus-host disease (GVHD). Donor APCs play a lesser role in the induction of acute GVHD despite their predicted capacity to cross-present host antigens. In contrast, donor APCs may play a role in perpetuating the tissue injury observed in chronic GVHD. Host APCs are also required for maximal graft-versus-leukemia responses. Recent studies have suggested potential strategies by which the continued presence of host APCs can be exploited to prime strong donor immunity to tumors without the induction of GVHD.
Collapse
Affiliation(s)
- Ronjon Chakraverty
- Department of Hematology, University College London, London, United Kingdom
| | | |
Collapse
|
32
|
Durakovic N, Radojcic V, Skarica M, Bezak KB, Powell JD, Fuchs EJ, Luznik L. Factors governing the activation of adoptively transferred donor T cells infused after allogeneic bone marrow transplantation in the mouse. Blood 2007; 109:4564-74. [PMID: 17227829 PMCID: PMC1885486 DOI: 10.1182/blood-2006-09-048124] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Murine models of bone marrow transplantation were used to study the mechanisms governing the activation of donor lymphocyte infusions (DLIs) manifesting as lymphohematopoietic graft-versus-host (LH-GVH) and graft-versus-leukemia (GVL) reactivities. We demonstrate here that established mixed chimerism influences the potency of DLI-mediated alloreactivity only in the MHC-mismatched but not MHC-matched setting. In the MHC-matched setting, high levels (>or= 40%) of residual host chimerism correlated negatively with DLI-mediated alloreactivity irrespective of the timing of their administration, the donor's previous sensitization to host antigens, or the level of residual host APCs. In vivo administration of Toll-like receptor (TLR) ligands was required to maximize DLI-mediated LH-GVH and GVL reactivities in chimeras with low levels (<or= 15%) of residual host chimerism. In contrast, coadministration of DLI with antigen-presenting cell (APC) activators was insufficient to augment their LH-GVH response in the presence of high levels of host chimerism unless the host's T cells were transiently depleted. Together, these results show the cardinal influence of donor-host incompatibility on DLI-mediated GVH responses and suggest that in MHC-matched chimeras, the induction of optimal alloreactivity requires not only donor T cells and host APCs but also TLR ligands and in the presence of high levels of host chimerism depletion of host T cells.
Collapse
Affiliation(s)
- Nadira Durakovic
- Divisions of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|