201
|
Locke FL, Pidala J, Storer B, Martin PJ, Pulsipher MA, Chauncey TR, Jacobsen N, Kröger N, Walker I, Light S, Shaw BE, Beato F, Laport GG, Nademanee A, Keating A, Socie G, Anasetti C. CD25 Blockade Delays Regulatory T Cell Reconstitution and Does Not Prevent Graft-versus-Host Disease After Allogeneic Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2016; 23:405-411. [PMID: 28007665 DOI: 10.1016/j.bbmt.2016.12.624] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/14/2016] [Indexed: 11/26/2022]
Abstract
Daclizumab, a humanized monoclonal antibody, binds CD25 and blocks formation of the IL-2 receptor on T cells. A study of daclizumab as acute graft-versus-host disease (GVHD) prophylaxis after unrelated bone marrow transplantation was conducted before the importance of CD25+FOXP3+ regulatory T cells (Tregs) was recognized. Tregs can abrogate the onset of GVHD. The relation between Tregs and a graft-versus-malignancy effect is not fully understood. An international, multicenter, double-blind clinical trial randomized 210 adult or pediatric patients to receive 5 weekly doses of daclizumab at 0.3 mg/kg (n = 69) or 1.2 mg/kg (n = 76) or placebo (n = 65) after unrelated marrow transplantation for treatment of hematologic malignancies or severe aplastic anemia. The risk of acute GVHD did not differ among the groups (P = .68). Long-term follow-up of clinical outcomes and correlative analysis of peripheral blood T cell phenotype suggested that the patients treated with daclizumab had an increased risk of chronic GVHD (hazard ratio [HR], 1.49; 95% confidence interval [CI], 1.0 to 2.3; P = .08) and a decreased risk of relapse (HR, 0.57; 95% CI, 0.3 to 1.0; P = .05), but similar survival (HR, 0.89; 95% CI, 0.6 to 1.3; P = .53). T cells from a subset of patients (n = 107) were analyzed by flow cytometry. Compared with placebo, treatment with daclizumab decreased the proportion of Tregs among CD4 T cells at days 11-35 and increased the proportion of central memory cells among CD4 T cells at 1 year. Prophylactic administration of daclizumab does not prevent acute GVHD, but may increase the risk of chronic GVHD and decrease the risk of relapse. By delaying Treg reconstitution and promoting immunologic memory, anti-CD25 therapy may augment alloreactivity and antitumor immunity.
Collapse
Affiliation(s)
- Frederick L Locke
- Blood and Marrow Transplantation Program, Moffitt Cancer Center, Tampa, Florida
| | - Joseph Pidala
- Blood and Marrow Transplantation Program, Moffitt Cancer Center, Tampa, Florida
| | - Barry Storer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Paul J Martin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, Division of Oncology, University of Washington, Seattle, Washington
| | - Michael A Pulsipher
- Blood and Marrow Transplant Program, Huntsman Cancer Institute, University of Utah Medical Center, Salt Lake City, Utah
| | - Thomas R Chauncey
- Department of Medicine, Division of Oncology, University of Washington, Seattle, Washington; Cancer Care Division, VA Puget Sound Health Care System, Seattle, Washington
| | - Niels Jacobsen
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irwin Walker
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Bronwen E Shaw
- Haemato-Oncology Unit, Royal Marsden Hospital, Sutton, United Kingdom
| | - Francisca Beato
- Blood and Marrow Transplantation Program, Moffitt Cancer Center, Tampa, Florida
| | - Ginna G Laport
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California
| | - Auayporn Nademanee
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope, Duarte, California
| | - Armand Keating
- Blood and Marrow Transplant Centre, Department of Hematology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Gerard Socie
- Department of Hematology/Transplantation, Hospital Saint Louis, Paris, France
| | - Claudio Anasetti
- Blood and Marrow Transplantation Program, Moffitt Cancer Center, Tampa, Florida.
| |
Collapse
|
202
|
Berges C, Kerkau T, Werner S, Wolf N, Winter N, Hünig T, Einsele H, Topp MS, Beyersdorf N. Hsp90 inhibition ameliorates CD4 + T cell-mediated acute Graft versus Host disease in mice. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:463-473. [PMID: 27980780 PMCID: PMC5134726 DOI: 10.1002/iid3.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 08/18/2016] [Accepted: 08/21/2016] [Indexed: 11/22/2022]
Abstract
Introduction For many patients with leukemia only allogeneic bone marrow transplantion provides a chance of cure. Co‐transplanted mature donor T cells mediate the desired Graft versus Tumor (GvT) effect required to destroy residual leukemic cells. The donor T cells very often, however, also attack healthy tissue of the patient inducing acute Graft versus Host Disease (aGvHD)—a potentially life‐threatening complication. Methods Therefore, we used the well established C57BL/6 into BALB/c mouse aGvHD model to evaluate whether pharmacological inhibition of heat shock protein 90 (Hsp90) would protect the mice from aGvHD. Results Treatment of the BALB/c recipient mice from day 0 to +2 after allogeneic CD4+ T cell transplantation with the Hsp90 inhibitor 17‐(dimethylaminoethylamino)‐17‐demethoxygeldanamycin (DMAG) partially protected the mice from aGvHD. DMAG treatment was, however, insufficient to prolong overall survival of leukemia‐bearing mice after transplantation of allogeneic CD4+ and CD8+ T cells. Ex vivo analyses and in vitro experiments revealed that DMAG primarily inhibits conventional CD4+ T cells with a relative resistance of CD4+ regulatory and CD8+ T cells toward Hsp90 inhibition. Conclusions Our data, thus, suggest that Hsp90 inhibition might constitute a novel approach to reduce aGvHD in patients without abrogating the desired GvT effect.
Collapse
Affiliation(s)
- Carsten Berges
- Department of Internal Medicine II Division of Hematology University Hospital Würzburg Würzburg Germany
| | - Thomas Kerkau
- Institute for Virology and Immunobiology University of Würzburg Würzburg Germany
| | - Sandra Werner
- Institute for Virology and Immunobiology University of Würzburg Würzburg Germany
| | - Nelli Wolf
- Institute for Virology and Immunobiology University of Würzburg Würzburg Germany
| | - Nadine Winter
- Department of Internal Medicine II Division of Hematology University Hospital Würzburg Würzburg Germany
| | - Thomas Hünig
- Institute for Virology and Immunobiology University of Würzburg Würzburg Germany
| | - Hermann Einsele
- Department of Internal Medicine II Division of Hematology University Hospital Würzburg Würzburg Germany
| | - Max S Topp
- Department of Internal Medicine II Division of Hematology University Hospital Würzburg Würzburg Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology University of Würzburg Würzburg Germany
| |
Collapse
|
203
|
Belle L, Fransolet G, Somja J, Binsfeld M, Delvenne P, Drion P, Hannon M, Beguin Y, Ehx G, Baron F. Limited Impact of Imatinib in a Murine Model of Sclerodermatous Chronic Graft-versus-Host Disease. PLoS One 2016; 11:e0167997. [PMID: 27942010 PMCID: PMC5152855 DOI: 10.1371/journal.pone.0167997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 11/23/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Sclerodermatous chronic Graft-versus-Host Disease (scl-cGVHD) is one of the most severe form of cGVHD. The Platelet-derived Grotwth Factor (PDGF) and the Transforming Growth Factor-β (TGF-β) play a significant role in the fibrosing process occurring in scl-cGVHD. This prompted us to assess the impact of the PDGF-r and c-Abl tyrosine kinase inhibitor imatinib on scl-cGVHD. METHODS To assess the impact of imatinib on T cell subset proliferation in vivo, Balb/cJ recipient mice were lethally (7 Gy) irradiated and then injected with 10x106 bone marrow cells from B10.D2 mice on day 0. Fourteen days later, 70x106 carboxyfluorescein succinimidyl ester (CFSE)-labeled splenocytes from B10.D2 mice were infused and imatinib or sterile water was administered for 5 days. To induce severe scl-cGVHD, Balb/cJ mice were injected i.v. with 10.106 bone marrow cells and 70.106 splenocytes from B10.D2 donor mice after 7 Gy irradiation. Mice were then given sterile water or imatinib from day +7 after transplantation to the end of the experiment (day +52). RESULTS Imatinib decreased the proliferation of total T cells (P = 0.02), CD8+ T cells (P = 0.01), and of regulatory T cells (Tregs) (P = 0.02) in the spleen. In the severe scl-cGVHD model, imatinib-treated mice had significantly lower levels of PDGF-r phosphorylation than control mice on day 29 after transplantation (P = 0.008). However, scl-cGVHD scores were similar between vehicle- and imatinib-treated mice during the whole experiment, while there was a suggestion for less weight loss in imatinib-treated mice that reached statistical significance at day +52 following transplantation (P = 0.02). CONCLUSIONS Imatinib had a limited impact in murine scl-cGVHD despite significant inhibition of PDGF-r.
Collapse
Affiliation(s)
- Ludovic Belle
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Gilles Fransolet
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- * E-mail:
| | - Joan Somja
- Department of Pathology, University of Liège, Liège, Belgium
| | - Marilène Binsfeld
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | | | | | - Muriel Hannon
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Yves Beguin
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- Department of Medicine, Division of Hematology, CHU of Liège, Liège, Belgium
| | - Grégory Ehx
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Frédéric Baron
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- Department of Medicine, Division of Hematology, CHU of Liège, Liège, Belgium
| |
Collapse
|
204
|
|
205
|
Pharmacodynamics of T cell function for monitoring pharmacologic immunosuppression after allogeneic hematopoietic stem cell transplantation. Int J Hematol 2016; 105:497-505. [PMID: 27882485 DOI: 10.1007/s12185-016-2145-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 11/10/2016] [Accepted: 11/10/2016] [Indexed: 01/20/2023]
Abstract
Information on pharmacodynamic monitoring after allogeneic hematopoietic cell transplantation (allo-SCT) to evaluate individual responses to immunosuppressive drugs is scarce. We studied the relationship between a panel of pharmacodynamic markers monitored during the first 3 months after transplant and the occurrence of graft-versus-host disease (GVHD). Lymphocyte activation assessed by intracellular ATP concentration in CD4+ T cells, a high percentage of CD8+ effector T cells, and a low percentage of CD4+ regulatory T (Treg) cells correlated significantly with GVHD. A cutoff value of 0.5 for the CD8+ effector T/Treg ratio provided the most accurate diagnosis of GVHD (sensitivity 58.8%, specificity 91%). These pharmacodynamic markers may provide an efficient complement to standard pharmacokinetic monitoring of immunosuppressive drugs after allo-SCT.
Collapse
|
206
|
Al-Homsi AS, Goodyke A, McLane M, Abdel-Mageed S, Cole K, Muilenburg M, Feng Y. Post-Transplantation Cyclophosphamide and Ixazomib Combination Rescues Mice Subjected to Experimental Graft-versus-Host Disease and Is Superior to Either Agent Alone. Biol Blood Marrow Transplant 2016; 23:255-261. [PMID: 27888016 DOI: 10.1016/j.bbmt.2016.11.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/16/2016] [Indexed: 01/13/2023]
Abstract
Lapses in the prevention of graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT) warrant novel approaches. Such approaches include, among others, the use of post-transplantation cyclophosphamide (PTC) and proteasome inhibitors. Although PTC alone consistently produces low rates of chronic GVHD, the incidence of acute GVHD remains significant. Inversely, prolonged post-transplantation administration of proteasome inhibitors carries a risk of paradoxical aggravation of GVHD. We examined whether the combination of cyclophosphamide and ixazomib addresses the limitations of each of these agents when used alone to prevent GVHD in mice subjected to allogeneic HSCT across MHC barriers. We chose ixazomib, an orally bioavailable proteasome inhibitor, because of its favorable physiochemical characteristics. The combination of cyclophosphamide and ixazomib improved overall survival of mice in comparison to an untreated control group and to groups receiving either cyclophosphamide alone or ixazomib alone. Furthermore, cyclophosphamide prevented the surge of IL-1β, GVHD aggravation, and sudden death associated with prolonged administration of ixazomib after HSCT. Finally, we demonstrated that although ixazomib was administered before cyclophosphamide, it did not impair the preferential depletion of proliferating as opposed to resting donor T cells. Our data suggest that the combination of cyclophosphamide and ixazomib for the prevention of GVHD after allogeneic HSCT is promising and merits further investigation in clinical trials.
Collapse
Affiliation(s)
- Ahmad Samer Al-Homsi
- Adult Blood and Marrow Transplantation, Spectrum Health, Grand Rapids, Michigan; Michigan State University College of Human Medicine, Grand Rapids, Michigan.
| | - Austin Goodyke
- Adult Blood and Marrow Transplantation, Spectrum Health, Grand Rapids, Michigan
| | - Michael McLane
- Adult Blood and Marrow Transplantation, Spectrum Health, Grand Rapids, Michigan
| | - Sarah Abdel-Mageed
- Adult Blood and Marrow Transplantation, Spectrum Health, Grand Rapids, Michigan
| | - Kelli Cole
- Adult Blood and Marrow Transplantation, Spectrum Health, Grand Rapids, Michigan
| | - Marlee Muilenburg
- Adult Blood and Marrow Transplantation, Spectrum Health, Grand Rapids, Michigan
| | - Yuxin Feng
- Adult Blood and Marrow Transplantation, Spectrum Health, Grand Rapids, Michigan
| |
Collapse
|
207
|
LRBA is Essential for Allogeneic Responses in Bone Marrow Transplantation. Sci Rep 2016; 6:36568. [PMID: 27824136 PMCID: PMC5099895 DOI: 10.1038/srep36568] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/18/2016] [Indexed: 12/25/2022] Open
Abstract
The PH-BEACH-WD40 (PBW) protein family members play a role in coordinating receptor signaling and intracellular vesicle trafficking. LPS-Responsive-Beige-like Anchor (LRBA) is a PBW protein whose immune function remains elusive. Here we show that LRBA-null mice are viable, but exhibit compromised rejection of allogeneic, xenogeneic and missing self bone-marrow grafts. Further, we demonstrate that LRBA-null Natural Killer (NK) cells exhibit impaired signaling by the key NK activating receptors, NKp46 and NKG2D. However, induction of IFN-γ by cytokines remains intact, indicating LRBA selectively facilitates signals by receptors for ligands expressed on the surface of NK targets. Surprisingly, LRBA limits immunoregulatory cell numbers in tissues where GvHD is primed or initiated, and consistent with this LRBA-null mice also demonstrate resistance to lethal GvHD. These findings demonstrate that LRBA is redundant for host longevity while being essential for both host and donor-mediated immune responses and thus represents a unique and novel molecular target in transplant immunology.
Collapse
|
208
|
Mechanistic approaches for the prevention and treatment of chronic GVHD. Blood 2016; 129:22-29. [PMID: 27821505 DOI: 10.1182/blood-2016-08-686659] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023] Open
Abstract
Clinical outcomes for patients undergoing allogeneic hematopoietic stem cell transplantation continue to improve, but chronic graft-versus-host disease (GVHD) remains a common toxicity and major cause of nonrelapse morbidity and mortality. Treatment of chronic GVHD has previously relied primarily on corticosteroids and other broadly immune suppressive agents. However, conventional immune suppressive agents have limited clinical efficacy in chronic GVHD, and prolonged immune suppressive treatments result in additional toxicities that further limit clinical recovery from transplant and return to normal daily function. Recent advances in our understanding of the immune pathology of chronic GVHD offer the possibility that new therapeutic approaches can be directed in more precise ways to target specific immunologic mechanisms and pathways. In this review, we briefly summarize current standard treatment options and present new therapeutic approaches that are supported by preclinical studies and early-phase clinical trials suggesting that these approaches may have clinical utility for treatment or prevention of chronic GVHD. Further evaluation of these new therapeutic options in well-designed prospective multicenter trials are needed to identify the most effective new agents and improve outcomes for patients with chronic GVHD.
Collapse
|
209
|
Chronic graft-versus-host disease: biological insights from preclinical and clinical studies. Blood 2016; 129:13-21. [PMID: 27821504 DOI: 10.1182/blood-2016-06-686618] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/06/2016] [Indexed: 12/15/2022] Open
Abstract
With the increasing use of mismatched, unrelated, and granulocyte colony-stimulating factor-mobilized peripheral blood stem cell donor grafts and successful treatment of older recipients, chronic graft-versus-host disease (cGVHD) has emerged as the major cause of nonrelapse mortality and morbidity. cGVHD is characterized by lichenoid changes and fibrosis that affects a multitude of tissues, compromising organ function. Beyond steroids, effective treatment options are limited. Thus, new strategies to both prevent and treat disease are urgently required. Over the last 5 years, our understanding of cGVHD pathogenesis and basic biology, born out of a combination of mouse models and correlative clinical studies, has radically improved. We now understand that cGVHD is initiated by naive T cells, differentiating predominantly within highly inflammatory T-helper 17/T-cytotoxic 17 and T-follicular helper paradigms with consequent thymic damage and impaired donor antigen presentation in the periphery. This leads to aberrant T- and B-cell activation and differentiation, which cooperate to generate antibody-secreting cells that cause the deposition of antibodies to polymorphic recipient antigens (ie, alloantibody) or nonpolymorphic antigens common to both recipient and donor (ie, autoantibody). It is now clear that alloantibody can, in concert with colony-stimulating factor 1 (CSF-1)-dependent donor macrophages, induce a transforming growth factor β-high environment locally within target tissue that results in scleroderma and bronchiolitis obliterans, diagnostic features of cGVHD. These findings have yielded a raft of potential new therapeutics, centered on naive T-cell depletion, interleukin-17/21 inhibition, kinase inhibition, regulatory T-cell restoration, and CSF-1 inhibition. This new understanding of cGVHD finally gives hope that effective therapies are imminent for this devastating transplant complication.
Collapse
|
210
|
Fuji S, Shindo T. Friend or foe? Mogamulizumab in allogeneic hematopoietic stem cell transplantation for adult T-cell leukemia/lymphoma. Stem Cell Investig 2016; 3:70. [PMID: 27868052 DOI: 10.21037/sci.2016.09.13] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/13/2016] [Indexed: 12/28/2022]
Abstract
Adult T-cell leukemia/lymphoma (ATL/ATLL) is a peripheral T-cell neoplasm associated with human T-lymphotropic virus type-1 (HTLV-1). Even the currently most intensive chemotherapy regimen modified LSG15 (mLSG15, VCAP-AMP-VECP) results in a dismal clinical outcome, with a median overall survival of only around 1 year. Although allogeneic hematopoietic stem cell transplantation (allo-HSCT) may lead to long-term remission in a proportion of patients with aggressive ATL, the clinical outcome in patients with refractory or relapsed ATL is unsatisfactory. The anti-CCR4 antibody mogamulizumab (moga) has been recently approved for ATL in Japan, and it is effective in a significant proportion of patients with refractory or relapsed ATL. However, there are major concerns about the harmful influences of pretransplant moga on the immune reconstitution after allo-HSCT. Specifically, moga depletes regulatory T cells (Tregs) for at least a few months, which may increase the risk of graft-versus-host disease (GVHD) after allo-HSCT. A recent retrospective study from Japan clearly showed that pretransplant moga increased the risk of severe and steroid-refractory GVHD, which led to increases in non-relapse mortality and overall mortality. To improve the overall clinical outcome in patients with relapsed or refractory ATL, more studies are needed to incorporate moga without increasing adverse effects on the clinical outcome after allo-HSCT. In this review, we aim to provide an updated summary of the research related to moga and allo-HSCT.
Collapse
Affiliation(s)
- Shigeo Fuji
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Takero Shindo
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| |
Collapse
|
211
|
|
212
|
DR3 signaling modulates the function of Foxp3+ regulatory T cells and the severity of acute graft-versus-host disease. Blood 2016; 128:2846-2858. [PMID: 27760760 DOI: 10.1182/blood-2016-06-723783] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/29/2016] [Indexed: 12/20/2022] Open
Abstract
CD4+Foxp3+ regulatory T cells (Treg) are a subpopulation of T cells, which regulate the immune system and enhance immune tolerance after transplantation. Donor-derived Treg prevent the development of lethal acute graft-versus-host disease (GVHD) in murine models of allogeneic hematopoietic stem cell transplantation. We recently demonstrated that a single treatment of the agonistic antibody to DR3 (death receptor 3, αDR3) to donor mice resulted in the expansion of donor-derived Treg and prevented acute GVHD, although the precise role of DR3 signaling in GVHD has not been elucidated. In this study, we comprehensively analyzed the immunophenotype of Treg after DR3 signal activation, demonstrating that DR3-activated Treg (DR3-Treg) had an activated/mature phenotype. Furthermore, the CD25+Foxp3+ subpopulation in DR3-Treg showed stronger suppressive effects in vivo. Prophylactic treatment of αDR3 to recipient mice expanded recipient-derived Treg and reduced the severity of GVHD, whereas DR3 activation in mice with ongoing GVHD further promoted donor T-cell activation/proliferation. These data suggest that the function of DR3 signaling was highly dependent on the activation status of the T cells. In conclusion, our data demonstrated that DR3 signaling affects the function of Treg and T-cell activation after alloantigen exposure in a time-dependent manner. These observations provide important information for future clinical testing using human DR3 signal modulation and highlight the critical effect of the state of T-cell activation on clinical outcomes after activation of DR3.
Collapse
|
213
|
Wang Y, Zhao XY, Xu LP, Zhang XH, Han W, Chen H, Wang FR, Mo XD, Zhang YY, Zhao XS, Y K, Liua KY, Huang XJ, Yu XZ, Chang YJ. Lower incidence of acute GVHD is associated with the rapid recovery of CD4 +CD25 +CD45RA + regulatory T cells in patients who received haploidentical allografts from NIMA-mismatched donors: A retrospective (development) and prospective (validation) cohort-based study. Oncoimmunology 2016; 5:e1242546. [PMID: 28180031 DOI: 10.1080/2162402x.2016.1242546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 10/20/2022] Open
Abstract
To investigate the effects of non-inherited maternal antigen (NIMA) on clinical outcomes and immune recovery, especially of regulatory T cells (Tregs), in patients who underwent unmanipulated haploidentical transplantation. A retrospective cohort (n = 57) and a prospective cohort (n = 88) were included. All patients received haploidentical allografts from sibling donors. Reconstitution of immune subsets, including Tregs, was determined using multicolor flow cytometry. In the retrospective cohort, the cumulative incidence of grades II-IV acute GVHD in patients with NIMA-mismatched donors was significantly lower than that of cases with NIPA-mismatched donors (14.8% vs. 43.30%, p = 0.018). Patients with higher percentages of CD4+CD25+CD45RA+ T cells (naive Tregs) within CD4+ T cells recovered on day 30 (≥1.55%) experienced a significantly lower incidence of grades II-IV acute GVHD than that of cases with lower percentages of naive Tregs (<1.55%) (13.8% vs. 46.4%, p = 0.010). Multivariate analysis showed that NIMA mismatch and the percentages of naive Tregs were associated with the incidence of grades II-IV acute GVHD [p = 0.050, and 0.031, respectively]. In the prospective cohort, the association of NIMA mismatch [HR = 0.365, 95% CI, 0.169-0.786, p = 0.010] or higher percentages of naive Tregs recovered on day 30 (≥1.55%) [HR = 0.114, 95% CI, 0.027-0.479, p = 0.003] with a lower cumulative incidence of grades II-IV acute GVHD was further demonstrated. No effects of NIMA mismatch on chronic GVHD, transplant-related mortality, relapse, disease-free survival, or overall survival were found. Our results confirmed the role of NIMA mismatch in acute GVHD and provided the first demonstration, based on clinical data, that recovered Tregs may be involved in the effects of NIMA on acute GVHD in a haploidentical transplant setting.
Collapse
Affiliation(s)
- Yu Wang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Xiang-Yu Zhao
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Lan-Ping Xu
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Xiao-Hui Zhang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Wei Han
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Huan Chen
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Feng-Rong Wang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Xiao-Dong Mo
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Yuan-Yuan Zhang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Xiao-Su Zhao
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Kong Y
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Kai-Yan Liua
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, P.R. China; Peking-Tsinghua Center for Life Sciences, Beijing, P.R. China
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA; Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation , Beijing, P.R. China
| |
Collapse
|
214
|
Perobelli SM, Mercadante ACT, Galvani RG, Gonçalves-Silva T, Alves APG, Pereira-Neves A, Benchimol M, Nóbrega A, Bonomo A. G-CSF-Induced Suppressor IL-10+ Neutrophils Promote Regulatory T Cells That Inhibit Graft-Versus-Host Disease in a Long-Lasting and Specific Way. THE JOURNAL OF IMMUNOLOGY 2016; 197:3725-3734. [PMID: 27707998 DOI: 10.4049/jimmunol.1502023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 09/06/2016] [Indexed: 01/13/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is the main complication of allogeneic hematopoietic stem cell transplantation, and many efforts have been made to overcome this important limitation. We showed previously that G-CSF treatment generates low-density splenic granulocytes that inhibit experimental aGVHD. In this article, we show that aGVHD protection relies on incoming IL-10+ neutrophils from G-CSF-treated donor spleen (G-Neutrophils). These G-Neutrophils have high phagocytic capacity, high peroxide production, low myeloperoxidase activity, and low cytoplasmic granule content, which accounts for their low density. Furthermore, they have low expression of MHC class II, costimulatory molecules, and low arginase1 expression. Also, they have low IFN-γ, IL-17F, IL-2, and IL-12 levels, with increased IL-10 production and NO synthase 2 expression. These features are in accordance with the modulatory capacity of G-Neutrophils on regulatory T cell (Treg) generation. In vivo, CD25+ Treg depletion shortly after transplantation with splenic cells from G-CSF-treated donors blocks suppression of aGVHD, suggesting Treg involvement in the protection induced by the G-Neutrophils. The immunocompetence and specificity of the semiallogeneic T cells, long-term after the bone marrow transplant using G-Neutrophils, were confirmed by third-party skin graft rejection; importantly, a graft-versus-leukemia assay showed that T cell activity was maintained, and all of the leukemic cells were eliminated. We conclude that G-CSF treatment generates a population of activated and suppressive G-Neutrophils that reduces aGVHD in an IL-10- and Treg-dependent manner, while maintaining immunocompetence and the graft versus leukemia effect.
Collapse
Affiliation(s)
- Suelen Martins Perobelli
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil.,Divisão de Medicina Experimental, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil.,Laboratório de Pesquisa Sobre o Timo, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | | | - Rômulo Gonçalves Galvani
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil.,Divisão de Medicina Experimental, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil.,Laboratório de Pesquisa Sobre o Timo, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | - Triciana Gonçalves-Silva
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil.,Divisão de Medicina Experimental, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil.,Laboratório de Pesquisa Sobre o Timo, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | - Ana Paula Gregório Alves
- Divisão de Medicina Experimental, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil
| | - Antonio Pereira-Neves
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil.,Departamento de Microbiologia, FIOCRUZ Pernambuco, Pernambuco 50670-420, Brazil
| | - Marlene Benchimol
- Universidade do Grande, Rio de Janeiro 25071-202, Brazil.,Instituto Nacional de Biologia Estrutural e Bioimagem da Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil; and
| | - Alberto Nóbrega
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil
| | - Adriana Bonomo
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil; .,Divisão de Medicina Experimental, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil.,Laboratório de Pesquisa Sobre o Timo, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, Brazil.,Programa FIOCancer, Vice Presidência de Pesquisa e Laboratórios de Referência/FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| |
Collapse
|
215
|
Martelli MF, Aversa F. Haploidentical transplants using ex vivo T-cell depletion. Semin Hematol 2016; 53:252-256. [DOI: 10.1053/j.seminhematol.2016.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/20/2016] [Indexed: 02/02/2023]
|
216
|
Edinur HA, Manaf SM, Che Mat NF. Genetic barriers in transplantation medicine. World J Transplant 2016; 6:532-541. [PMID: 27683631 PMCID: PMC5036122 DOI: 10.5500/wjt.v6.i3.532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/26/2016] [Accepted: 07/13/2016] [Indexed: 02/05/2023] Open
Abstract
The successful of transplantation is determined by the shared human leukocyte antigens (HLAs) and ABO blood group antigens between donor and recipient. In recent years, killer cell receptor [i.e., killer cell immunoglobulin-like receptor (KIR)] and major histocompatibility complex (MHC) class I chain-related gene molecule (i.e., MICA) were also reported as important determinants of transplant compatibility. At present, several different genotyping techniques (e.g., sequence specific primer and sequence based typing) can be used to characterize blood group, HLA, MICA and KIR and loci. These molecular techniques have several advantages because they do not depend on the availability of anti-sera, cellular expression and have greater specificity and accuracy compared with the antibody-antigen based typing. Nonetheless, these molecular techniques have limited capability to capture increasing number of markers which have been demonstrated to determine donor and recipient compatibility. It is now possible to genotype multiple markers and to the extent of a complete sequencing of the human genome using next generation sequencer (NGS). This high throughput genotyping platform has been tested for HLA, and it is expected that NGS will be used to simultaneously genotype a large number of clinically relevant transplantation genes in near future. This is not far from reality due to the bioinformatics support given by the immunogenetics community and the rigorous improvement in NGS methodology. In addition, new developments in immune tolerance based therapy, donor recruitment strategies and bioengineering are expected to provide significant advances in the field of transplantation medicine.
Collapse
|
217
|
Le Texier L, Lineburg KE, Cao B, McDonald-Hyman C, Leveque-El Mouttie L, Nicholls J, Melino M, Nalkurthi BC, Alexander KA, Teal B, Blake SJ, Souza-Fonseca-Guimaraes F, Engwerda CR, Kuns RD, Lane SW, Teng M, Teh C, Gray D, Clouston AD, Nilsson SK, Blazar BR, Hill GR, MacDonald KP. Autophagy-dependent regulatory T cells are critical for the control of graft-versus-host disease. JCI Insight 2016; 1:e86850. [PMID: 27699243 DOI: 10.1172/jci.insight.86850] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Regulatory T cells (Tregs) play a crucial role in the maintenance of peripheral tolerance. Quantitative and/or qualitative defects in Tregs result in diseases such as autoimmunity, allergy, malignancy, and graft-versus-host disease (GVHD), a serious complication of allogeneic stem cell transplantation (SCT). We recently reported increased expression of autophagy-related genes (Atg) in association with enhanced survival of Tregs after SCT. Autophagy is a self-degradative process for cytosolic components that promotes cell homeostasis and survival. Here, we demonstrate that the disruption of autophagy within FoxP3+ Tregs (B6.Atg7fl/fl-FoxP3cre+ ) resulted in a profound loss of Tregs, particularly within the bone marrow (BM). This resulted in dysregulated effector T cell activation and expansion, and the development of enterocolitis and scleroderma in aged mice. We show that the BM compartment is highly enriched in TIGIT+ Tregs and that this subset is differentially depleted in the absence of autophagy. Moreover, following allogeneic SCT, recipients of grafts from B6.Atg7fl/fl-FoxP3cre+ donors exhibited reduced Treg reconstitution, exacerbated GVHD, and reduced survival compared with recipients of B6.WT-FoxP3cre+ grafts. Collectively, these data indicate that autophagy-dependent Tregs are critical for the maintenance of tolerance after SCT and that the promotion of autophagy represents an attractive immune-restorative therapeutic strategy after allogeneic SCT.
Collapse
Affiliation(s)
- Laëtitia Le Texier
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katie E Lineburg
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Benjamin Cao
- Manufacturing, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Cameron McDonald-Hyman
- Pediatric Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lucie Leveque-El Mouttie
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jemma Nicholls
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Michelle Melino
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Blessy C Nalkurthi
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kylie A Alexander
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bianca Teal
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Stephen J Blake
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Christian R Engwerda
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Rachel D Kuns
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Steven W Lane
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Department of Bone Marrow Transplantation, Royal Brisbane Hospital, Brisbane, Queensland, Australia
| | - Michele Teng
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Charis Teh
- Molecular Genetics of Cancer Division and Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Daniel Gray
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.,Molecular Genetics of Cancer Division and Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | | | - Susan K Nilsson
- Manufacturing, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Bruce R Blazar
- Pediatric Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geoffrey R Hill
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Department of Bone Marrow Transplantation, Royal Brisbane Hospital, Brisbane, Queensland, Australia
| | - Kelli Pa MacDonald
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
218
|
Zeiser R, Socié G, Blazar BR. Pathogenesis of acute graft-versus-host disease: from intestinal microbiota alterations to donor T cell activation. Br J Haematol 2016; 175:191-207. [PMID: 27619472 DOI: 10.1111/bjh.14295] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 06/30/2016] [Accepted: 06/30/2016] [Indexed: 01/03/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a major life-threatening complication of allogeneic haematopoietic cell transplantation (allo-HCT). Here we discuss the aGVHD pathophysiology initiated by multiple signals that cause alloreactive T-cell activation. The outcome of such donor T-cell activation is influenced by T-cell receptor-signal strength, anatomical location, co-stimulatory/co-inhibitory signals and differentiation stage (naive, effector/memory) of T-cells. Additionally, cross-priming of T cells to antigens expressed by pathogens can contribute to aGVHD-mediated tissue injury. In addition to the properties of donor T-cell activation, highly specialized tissue resident cell types, such as innate lymphoid cells, antigen-presenting cells, immune regulatory cells and various intestinal cell populations are critically involved in aGVHD pathogenesis. The role of the thymus and secondary lymphoid tissue injury, non-haematopoietic cells, intestinal microflora, cytokines, chemokines, microRNAs, metabolites and kinases in aGVHD pathophysiology will be highlighted. Acute GVHD pathogenic mechanisms will be connected to novel therapeutic approaches under development for, and tested in, the clinic.
Collapse
Affiliation(s)
- Robert Zeiser
- Department of Haematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Centre, Freiburg, Germany.
| | - Gerard Socié
- Haematology Stem cell transplant Unit, Saint Louis Hospital, APHP, Paris, France
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
219
|
Boieri M, Shah P, Dressel R, Inngjerdingen M. The Role of Animal Models in the Study of Hematopoietic Stem Cell Transplantation and GvHD: A Historical Overview. Front Immunol 2016; 7:333. [PMID: 27625651 PMCID: PMC5003882 DOI: 10.3389/fimmu.2016.00333] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022] Open
Abstract
Bone marrow transplantation (BMT) is the only therapeutic option for many hematological malignancies, but its applicability is limited by life-threatening complications, such as graft-versus-host disease (GvHD). The last decades have seen great advances in the understanding of BMT and its related complications; in particular GvHD. Animal models are beneficial to study complex diseases, as they allow dissecting the contribution of single components in the development of the disease. Most of the current knowledge on the therapeutic mechanisms of BMT derives from studies in animal models. Parallel to BMT, the understanding of the pathophysiology of GvHD, as well as the development of new treatment regimens, has also been supported by studies in animal models. Pre-clinical experimentation is the basis for deep understanding and successful improvements of clinical applications. In this review, we retrace the history of BMT and GvHD by describing how the studies in animal models have paved the way to the many advances in the field. We also describe how animal models contributed to the understanding of GvHD pathophysiology and how they are fundamental for the discovery of new treatments.
Collapse
Affiliation(s)
- Margherita Boieri
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Marit Inngjerdingen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
220
|
Tsubokura Y, Satake A, Hotta M, Yoshimura H, Fujita S, Azuma Y, Nakanishi T, Nakaya A, Ito T, Ishii K, Nomura S. Successful treatment with mogamulizumab followed by allogeneic hematopoietic stem-cell transplantation in adult T-cell leukemia/lymphoma: a report of two cases. Int J Hematol 2016; 104:744-748. [PMID: 27573760 DOI: 10.1007/s12185-016-2087-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 01/25/2023]
Abstract
A humanized anti-CC chemokine receptor 4 (CCR4) monoclonal antibody, mogamulizumab (MOG), has been shown to be safe and effective in the treatment of relapsed/refractory adult T-cell leukemia/lymphoma (ATLL). MOG depletes ATLL cells as well as regulatory T cells (Tregs), as CCR4 is expressed on these cells as well. In this context, pretransplant treatment with MOG may induce severe graft-versus-host disease (GVHD) in allogeneic hematopoietic stem-cell transplantation (HSCT). However, the influence of MOG on allogeneic HSCT, including its induction of GVHD, is unclear. In this report, we describe two patients treated with MOG who subsequently underwent allogeneic HSCT. They did not develop severe GVHD or treatment-related complications. In addition, we examined the kinetics of Tregs in the second case. Finally, we suggest that the detrimental effects of MOG can be avoided, which should be prospectively evaluated in future studies.
Collapse
Affiliation(s)
- Yukie Tsubokura
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Atsushi Satake
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan.
| | - Masaaki Hotta
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Hideaki Yoshimura
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Shinya Fujita
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Yoshiko Azuma
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Takahisa Nakanishi
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Aya Nakaya
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Tomoki Ito
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Kazuyoshi Ishii
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| |
Collapse
|
221
|
Chopra M, Biehl M, Steinfatt T, Brandl A, Kums J, Amich J, Vaeth M, Kuen J, Holtappels R, Podlech J, Mottok A, Kraus S, Jordán-Garrote AL, Bäuerlein CA, Brede C, Ribechini E, Fick A, Seher A, Polz J, Ottmüller KJ, Baker J, Nishikii H, Ritz M, Mattenheimer K, Schwinn S, Winter T, Schäfer V, Krappmann S, Einsele H, Müller TD, Reddehase MJ, Lutz MB, Männel DN, Berberich-Siebelt F, Wajant H, Beilhack A. Exogenous TNFR2 activation protects from acute GvHD via host T reg cell expansion. J Exp Med 2016; 213:1881-900. [PMID: 27526711 PMCID: PMC4995078 DOI: 10.1084/jem.20151563] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/24/2016] [Indexed: 12/22/2022] Open
Abstract
Activation of TNFR2 with a novel agonist expands T reg cells in vivo and protects allo-HCT recipients from acute GvHD while sparing antilymphoma and antiinfectious properties of transplanted donor T cells. Donor CD4+Foxp3+ regulatory T cells (T reg cells) suppress graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (HCT [allo-HCT]). Current clinical study protocols rely on the ex vivo expansion of donor T reg cells and their infusion in high numbers. In this study, we present a novel strategy for inhibiting GvHD that is based on the in vivo expansion of recipient T reg cells before allo-HCT, exploiting the crucial role of tumor necrosis factor receptor 2 (TNFR2) in T reg cell biology. Expanding radiation-resistant host T reg cells in recipient mice using a mouse TNFR2-selective agonist before allo-HCT significantly prolonged survival and reduced GvHD severity in a TNFR2- and T reg cell–dependent manner. The beneficial effects of transplanted T cells against leukemia cells and infectious pathogens remained unaffected. A corresponding human TNFR2-specific agonist expanded human T reg cells in vitro. These observations indicate the potential of our strategy to protect allo-HCT patients from acute GvHD by expanding T reg cells via selective TNFR2 activation in vivo.
Collapse
Affiliation(s)
- Martin Chopra
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Marlene Biehl
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Tim Steinfatt
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Andreas Brandl
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Juliane Kums
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Jorge Amich
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Martin Vaeth
- Department of Molecular Pathology, Institute of Pathology, Würzburg University, 97080 Würzburg, Germany
| | - Janina Kuen
- Department of Molecular Pathology, Institute of Pathology, Würzburg University, 97080 Würzburg, Germany
| | - Rafaela Holtappels
- Institute for Virology and Research Center of Immunotherapy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Jürgen Podlech
- Institute for Virology and Research Center of Immunotherapy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Anja Mottok
- Institute of Pathology, Würzburg University, 97080 Würzburg, Germany
| | - Sabrina Kraus
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Ana-Laura Jordán-Garrote
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Carina A Bäuerlein
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Christian Brede
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Eliana Ribechini
- Institute for Virology and Immunobiology, Würzburg University, 97080 Würzburg, Germany
| | - Andrea Fick
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Axel Seher
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Johannes Polz
- Institute of Immunology, Regensburg University, 93053 Regensburg, Germany
| | - Katja J Ottmüller
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Jeanette Baker
- Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA 94305
| | - Hidekazu Nishikii
- Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA 94305
| | - Miriam Ritz
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Katharina Mattenheimer
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Stefanie Schwinn
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Thorsten Winter
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Viktoria Schäfer
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Sven Krappmann
- Microbiology Institute, Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Thomas D Müller
- Department for Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute, Würzburg University, 97080 Würzburg, Germany
| | - Matthias J Reddehase
- Institute for Virology and Research Center of Immunotherapy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, Würzburg University, 97080 Würzburg, Germany
| | - Daniela N Männel
- Institute of Immunology, Regensburg University, 93053 Regensburg, Germany
| | | | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| |
Collapse
|
222
|
Control of GVHD by regulatory T cells depends on TNF produced by T cells and TNFR2 expressed by regulatory T cells. Blood 2016; 128:1651-9. [PMID: 27506541 DOI: 10.1182/blood-2016-02-700849] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/27/2016] [Indexed: 12/13/2022] Open
Abstract
Therapeutic CD4(+)Foxp3(+) natural regulatory T cells (Tregs) can control experimental graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HCT) by suppressing conventional T cells (Tconvs). Treg-based therapies are currently tested in clinical trials with promising preliminary results in allo-HCT. Here, we hypothesized that as Tregs are capable of modulating Tconv response, it is likely that the inflammatory environment and particularly donor T cells are also capable of influencing Treg function. Indeed, previous findings in autoimmune diabetes revealed a feedback mechanism that renders Tconvs able to stimulate Tregs by a mechanism that was partially dependent on tumor necrosis factor (TNF). We tested this phenomenon during alloimmune response in our previously described model of GVHD protection using antigen specific Tregs. Using different experimental approaches, we observed that control of GVHD by Tregs was fully abolished by blocking TNF receptor type 2 (TNFR2) or by using TNF-deficient donor T cells or TNFR2-deficient Tregs. Thus, our results show that Tconvs exert a powerful modulatory activity on therapeutic Tregs and clearly demonstrate that the sole defect of TNF production by donor T cells was sufficient to completely abolish the Treg suppressive effect in GVHD. Importantly, our findings expand the understanding of one of the central components of Treg action, the inflammatory context, and support that targeting TNF/TNFR2 interaction represents an opportunity to efficiently modulate alloreactivity in allo-HCT to either exacerbate it for a powerful antileukemic effect or reduce it to control GVHD.
Collapse
|
223
|
Zhou V, Agle K, Chen X, Beres A, Komorowski R, Belle L, Taylor C, Zhu F, Haribhai D, Williams CB, Verbsky J, Blumenschein W, Sadekova S, Bowman E, Ballantyne C, Weaver C, Serody DA, Vincent B, Serody J, Cua DJ, Drobyski WR. A colitogenic memory CD4+ T cell population mediates gastrointestinal graft-versus-host disease. J Clin Invest 2016; 126:3541-55. [PMID: 27500496 DOI: 10.1172/jci80874] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 06/09/2016] [Indexed: 12/21/2022] Open
Abstract
Damage to the gastrointestinal tract is a major cause of morbidity and mortality in graft-versus-host disease (GVHD) and is attributable to T cell-mediated inflammation. In this work, we identified a unique CD4+ T cell population that constitutively expresses the β2 integrin CD11c and displays a biased central memory phenotype and memory T cell transcriptional profile, innate-like properties, and increased expression of the gut-homing molecules α4β7 and CCR9. Using several complementary murine GVHD models, we determined that adoptive transfer and early accumulation of β2 integrin-expressing CD4+ T cells in the gastrointestinal tract initiated Th1-mediated proinflammatory cytokine production, augmented pathological damage in the colon, and increased mortality. The pathogenic effect of this CD4+ T cell population critically depended on coexpression of the IL-23 receptor, which was required for maximal inflammatory effects. Non-Foxp3-expressing CD4+ T cells produced IL-10, which regulated colonic inflammation and attenuated lethality in the absence of functional CD4+Foxp3+ T cells. Thus, the coordinate expression of CD11c and the IL-23 receptor defines an IL-10-regulated, colitogenic memory CD4+ T cell subset that is poised to initiate inflammation when there is loss of tolerance and breakdown of mucosal barriers.
Collapse
|
224
|
Blockade of interleukin-27 signaling reduces GVHD in mice by augmenting Treg reconstitution and stabilizing Foxp3 expression. Blood 2016; 128:2068-2082. [PMID: 27488350 DOI: 10.1182/blood-2016-02-698241] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/25/2016] [Indexed: 02/07/2023] Open
Abstract
Reestablishment of competent regulatory pathways has emerged as a strategy to reduce the severity of graft-versus-host disease (GVHD), and recalibrate the effector and regulatory arms of the immune system. However, clinically feasible, cost-effective strategies that do not require extensive ex vivo cellular manipulation have remained elusive. In the current study, we demonstrate that inhibition of the interleukin-27p28 (IL-27p28) signaling pathway through antibody blockade or genetic ablation prevented lethal GVHD in multiple murine transplant models. Moreover, protection from GVHD was attributable to augmented global reconstitution of CD4+ natural regulatory T cells (nTregs), CD4+ induced Tregs (iTregs), and CD8+ iTregs, and was more potent than temporally concordant blockade of IL-6 signaling. Inhibition of IL-27p28 also enhanced the suppressive capacity of adoptively transferred CD4+ nTregs by increasing the stability of Foxp3 expression. Notably, blockade of IL-27p28 signaling reduced T-cell-derived-IL-10 production in conventional T cells; however, there was no corresponding effect in CD4+ or CD8+ Tregs, indicating that IL-27 inhibition had differential effects on IL-10 production and preserved a mechanistic pathway by which Tregs are known to suppress GVHD. Targeting of IL-27 therefore represents a novel strategy for the in vivo expansion of Tregs and subsequent prevention of GVHD without the requirement for ex vivo cellular manipulation, and provides additional support for the critical proinflammatory role that members of the IL-6 and IL-12 cytokine families play in GVHD biology.
Collapse
|
225
|
Hu R, Liu Y, Su M, Song Y, Rood D, Lai L. Transplantation of Donor-Origin Mouse Embryonic Stem Cell-Derived Thymic Epithelial Progenitors Prevents the Development of Chronic Graft-versus-Host Disease in Mice. Stem Cells Transl Med 2016; 6:121-130. [PMID: 28170174 PMCID: PMC5442732 DOI: 10.5966/sctm.2016-0012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 06/16/2016] [Indexed: 01/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many malignant and nonmalignant diseases. However, chronic graft-versus-host disease (cGVHD) remains a significant cause of late morbidity and mortality after allogeneic HSCT. cGVHD often manifests as autoimmune syndrome. Thymic epithelial cells (TECs) play a critical role in supporting negative selection and regulatory T-cell (Treg) generation. Studies have shown that damage in TECs is sufficient to induce cGVHD. We have previously reported that mouse embryonic stem cells (mESCs) can be selectively induced to generate thymic epithelial progenitors (TEPs) in vitro. When transplanted in vivo, mESC-TEPs further develop into TECs that support T-cell development. We show here that transplantation of donor-origin mESC-TEPs into cGVHD recipients induces immune tolerance to both donor and host antigens and prevents the development of cGVHD. This is associated with more TECs and Tregs. Our results suggest that embryonic stem cell-derived TEPs may offer a new tool to control cGVHD. Stem Cells Translational Medicine 2017;6:121-130.
Collapse
Affiliation(s)
- Rong Hu
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, USA
- Guizhou Medical University, Guizhou, People's Republic of China
| | - Yalan Liu
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, USA
- Guizhou Medical University, Guizhou, People's Republic of China
| | - Yinhong Song
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Debra Rood
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, USA
- University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
226
|
miR-146b antagomir-treated human Tregs acquire increased GVHD inhibitory potency. Blood 2016; 128:1424-35. [PMID: 27485827 DOI: 10.1182/blood-2016-05-714535] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/25/2016] [Indexed: 12/24/2022] Open
Abstract
CD4(+)CD25(+)FoxP3(+) thymic-derived regulatory T cells (tTregs) are indispensable for maintaining immune system equilibrium. Adoptive transfer of tTregs is an effective means of suppressing graft-versus-host disease (GVHD) in murine models and in early human clinical trials. Tumor necrosis factor receptor-associated factor 6 (TRAF6), an ubiquitin-conjugating enzyme that mediates nuclear factor κB (NF-κB) activation, plays an essential role in modulating regulatory T cell survival and function. MicroRNAs (miRNAs) are noncoding RNAs, which mediate RNA silencing and posttranscriptional gene repression. By performing comprehensive TaqMan Low Density Array miRNA assays, we identified 10 miRNAs differentially regulated in human tTreg compared with control T cells. One candidate, miR-146b, is preferentially and highly expressed in human naive tTregs compared with naive CD4 T cells. miRNA prediction software revealed that TRAF6 was the one of the top 10 scored mRNAs involved tTreg function with the highest probability as a potential miR-146b target. Antagomir-mediated knockdown of miRNA-146b, but not another miRNA-146 family member (miRNA-146a), enhanced TRAF6 expression. TRAF6, in turn, increases NF-κB activation, which is essential for tTreg function as well as Foxp3 protein and antiapoptotic gene expression, and downregulates proapoptotic gene expression. miR-146b knockdown increased the nuclear localization and expression of genes regulated by NF-κB, which was associated with enhanced tTreg survival, proliferation, and suppressive function measured in vitro and in vivo. TRAF6 inhibition had the opposite effects. We conclude that an miR-146b-TRAF6-NF-κB-FoxP3 signaling pathway restrains regulatory T cell survival, proliferation, and suppressor function. In vitro exposure of human tTregs to miR-146b antagomirs can be exploited to improve the clinical efficacy of human adoptive tTreg transfer in a GVHD setting.
Collapse
|
227
|
Lifshitz GV, Zhdanov DD, Lokhonina AV, Eliseeva DD, Lyssuck EY, Zavalishin IA, Bykovskaia SN. Ex vivo expanded regulatory T cells CD4 +CD25 +FoxP3 +CD127 Low develop strong immunosuppressive activity in patients with remitting-relapsing multiple sclerosis. Autoimmunity 2016; 49:388-396. [PMID: 27424664 DOI: 10.1080/08916934.2016.1199020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by defect in regulatory function of CD4+CD25+ T cells. We demonstrated difference in proportion of regulatory T cells CD4+CD25+FoxP3+CD127low (Tregs) within the same patients' relapse and remission. Proportion of peripheral Tregs (pTregs) dropped almost two times in the relapse compare to remission. Levels of pTregs in patients' remission were lower than in healthy donors. Suppressive ability of pTregs was decreased in MS patients compared to healthy donors. Injections of expanded ex vivo autologous Tregs (eTregs) could be helpful in bringing up the level of Tregs in patients' blood. We developed a simple method for ex vivo expansion of autologous Tregs within a short period of time. The final pool of cells consisted of 90-95% eTregs. When we started the culture with 10-20 × 106 CD4+ T cells, we yield 300-400 × 106 eTregs in a week. Expression of FoxP3 and Helios was calculated by two methods. Expanded ex vivo patients' and donors' Tregs were characterized by increased from three to five times expression of FoxP3, as well as almost doubled Helios expression. Peripheral Tregs in MS patients have decreased demethylation of FoxP3 gene promoter in comparison with donors. On the contrary, eTregs showed stable up-regulated demethylation without difference between MS patients and donors. MS patients' and donors' eTregs have much more suppressive ability than pTregs. Our data showed that eTregs can be applied as immunotherapy for MS patients and other autoimmune diseases if further investigated.
Collapse
Affiliation(s)
- Gelena V Lifshitz
- a Pirogov Russian National Research Medical University , Moscow , Russia
| | - Dmitry D Zhdanov
- a Pirogov Russian National Research Medical University , Moscow , Russia.,b Regenex LLC, Skolkovo Innovation Center , Moscow , Russia , and
| | - Anastasia V Lokhonina
- a Pirogov Russian National Research Medical University , Moscow , Russia.,b Regenex LLC, Skolkovo Innovation Center , Moscow , Russia , and
| | - Daria D Eliseeva
- b Regenex LLC, Skolkovo Innovation Center , Moscow , Russia , and.,c Neurology Scientific Research Institute , Moscow , Russia
| | - Elena Y Lyssuck
- a Pirogov Russian National Research Medical University , Moscow , Russia.,b Regenex LLC, Skolkovo Innovation Center , Moscow , Russia , and
| | | | - Svetlana N Bykovskaia
- a Pirogov Russian National Research Medical University , Moscow , Russia.,b Regenex LLC, Skolkovo Innovation Center , Moscow , Russia , and
| |
Collapse
|
228
|
Chen LC, Nicholson YT, Rosborough BR, Thomson AW, Raimondi G. A Novel mTORC1-Dependent, Akt-Independent Pathway Differentiates the Gut Tropism of Regulatory and Conventional CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:1137-47. [PMID: 27402696 DOI: 10.4049/jimmunol.1600696] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/08/2016] [Indexed: 12/26/2022]
Abstract
The vitamin A metabolite all-trans retinoic acid (ATRA) induces a gut-homing phenotype in activated CD4(+) conventional T cells (Tconv) by upregulating the integrin α4β7 and the chemokine receptor CCR9. We report that, in contrast to mouse Tconv, only ∼50% of regulatory T cells (Treg) upregulate CCR9 when stimulated by physiological levels of ATRA, even though Tconv and Treg express similar levels of the retinoic acid receptor (RAR). The resulting bimodal CCR9 expression is not associated with differences in the extent of their proliferation, level of Foxp3 expression, or affiliation with naturally occurring Treg or induced Treg in the circulating Treg pool. Furthermore, we find that exposure of Treg to the mechanistic target of rapamycin (mTOR) inhibitor rapamycin suppresses upregulation of both CCR9 and α4β7, an effect that is not evident with Tconv. This suggests that in Treg, ATRA-induced upregulation of CCR9 and α4β7 is dependent on activation of a mTOR signaling pathway. The involvement of mTOR is independent of Akt activity, because specific inhibition of Akt, pyruvate dehydrogenase kinase-1, or its downstream target glycogen synthase kinase-3 did not prevent CCR9 expression. Additionally, Rictor (mTOR complex [mTORC]2)-deficient Treg showed unaltered ability to express CCR9, whereas Raptor (mTORC1)-deficient Treg were unable to upregulate CCR9, suggesting the selective participation of mTORC1. These findings reveal a novel difference between ATRA signaling and chemokine receptor induction in Treg versus Tconv and provide a framework via which the migratory behavior of Treg versus Tconv might be regulated differentially for therapeutic purposes.
Collapse
Affiliation(s)
- Leo C Chen
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and
| | - Yawah T Nicholson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and
| | - Brian R Rosborough
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and
| | - Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Giorgio Raimondi
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and
| |
Collapse
|
229
|
Corruption of dendritic cell antigen presentation during acute GVHD leads to regulatory T-cell failure and chronic GVHD. Blood 2016; 128:794-804. [PMID: 27338097 DOI: 10.1182/blood-2015-11-680876] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 06/11/2016] [Indexed: 12/20/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a major cause of late mortality following allogeneic bone marrow transplantation (BMT) and is characterized by tissue fibrosis manifesting as scleroderma and bronchiolitis obliterans. The development of acute GVHD (aGVHD) is a powerful clinical predictor of subsequent cGVHD, suggesting that aGVHD may invoke the immunologic pathways responsible for cGVHD. In preclinical models in which sclerodermatous cGVHD develops after a preceding period of mild aGVHD, we show that antigen presentation within major histocompatibility complex (MHC) class II of donor dendritic cells (DCs) is markedly impaired early after BMT. This is associated with a failure of regulatory T-cell (Treg) homeostasis and cGVHD. Donor DC-restricted deletion of MHC class II phenocopied this Treg deficiency and cGVHD. Moreover, specific depletion of donor Tregs after BMT also induced cGVHD, whereas adoptive transfer of Tregs ameliorated it. These data demonstrate that the defect in Treg homeostasis seen in cGVHD is a causative lesion and is downstream of defective antigen presentation within MHC class II that is induced by aGVHD.
Collapse
|
230
|
Thymic and Postthymic Regulation of Naïve CD4(+) T-Cell Lineage Fates in Humans and Mice Models. Mediators Inflamm 2016; 2016:9523628. [PMID: 27313405 PMCID: PMC4904118 DOI: 10.1155/2016/9523628] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022] Open
Abstract
Our understanding of how thymocytes differentiate into many subtypes has been increased progressively in its complexity. At early life, the thymus provides a suitable microenvironment with specific combination of stromal cells, growth factors, cytokines, and chemokines to induce the bone marrow lymphoid progenitor T-cell precursors into single-positive CD4+ and CD8+ T effectors and CD4+CD25+ T-regulatory cells (Tregs). At postthymic compartments, the CD4+ T-cells acquire distinct phenotypes which include the classical T-helper 1 (Th1), T-helper 2 (Th2), T-helper 9 (Th9), T-helper 17 (Th17), follicular helper T-cell (Tfh), and induced T-regulatory cells (iTregs), such as the regulatory type 1 cells (Tr1) and transforming growth factor-β- (TGF-β-) producing CD4+ T-cells (Th3). Tregs represent only a small fraction, 5–10% in mice and 1-2% in humans, of the overall CD4+ T-cells in lymphoid tissues but are essential for immunoregulatory circuits mediating the inhibition and expansion of all lineages of T-cells. In this paper, we first provide an overview of the major cell-intrinsic developmental programs that regulate T-cell lineage fates in thymus and periphery. Next, we introduce the SV40 immortomouse as a relevant mice model for implementation of new approaches to investigate thymus organogenesis, CD4 and CD8 development, and thymus cells tumorogenesis.
Collapse
|
231
|
Heinrichs J, Bastian D, Veerapathran A, Anasetti C, Betts B, Yu XZ. Regulatory T-Cell Therapy for Graft-versus-host Disease. JOURNAL OF IMMUNOLOGY RESEARCH AND THERAPY 2016; 1:1-14. [PMID: 27722210 PMCID: PMC5049884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Graft-versus-host disease (GVHD) is a significant cause of non-relapse mortality after allogeneic hematopoietic cell transplantation (allo-HCT). Existing strategies to prevent and treat GVHD are incomplete, where a significant portion of allo-HCT recipients developed this complication. Despite this, one such therapy has emerged involving the use of regulatory T cells (Tregs) to control GVHD. The use of natural Tregs (nTregs) yielded positive pre-clinical results and are actively under investigation to reduce GVHD. However, broad application of this approach may require standardization of Treg expansion methods and dosing. Inducible Tregs (iTregs) can be seamlessly generated, but controversial pre-clinical findings and phenotype instability have hampered their translation into the clinic. Here, we review the current biological differences between nTregs and iTregs, as well as their effects on GVHD and graft-versus-leukemia (GVL) responses. We conclude by exploring the idea of combinational cellular therapies for the prevention of GVHD and preservation of GVL.
Collapse
Affiliation(s)
- Jessica Heinrichs
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Pathology and Cell Biology, College of Medicine, University of South Florida, USA
| | - David Bastian
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | | | - Claudio Anasetti
- Department of Blood & Marrow Translation, Moffitt Cancer Center, Tampa, FL, USA
| | - Brain Betts
- Department of Blood & Marrow Translation, Moffitt Cancer Center, Tampa, FL, USA
| | - Xue-Zhong Yu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
232
|
Villa NY, Rahman MM, McFadden G, Cogle CR. Therapeutics for Graft-versus-Host Disease: From Conventional Therapies to Novel Virotherapeutic Strategies. Viruses 2016; 8:85. [PMID: 27011200 PMCID: PMC4810275 DOI: 10.3390/v8030085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has a curative potential for many hematologic malignancies and blood diseases. However, the success of allo-HSCT is limited by graft-versus-host disease (GVHD), an immunological syndrome that involves inflammation and tissue damage mediated by donor lymphocytes. Despite immune suppression, GVHD is highly incident even after allo-HSCT using human leukocyte antigen (HLA)-matched donors. Therefore, alternative and more effective therapies are needed to prevent or control GVHD while preserving the beneficial graft-versus-cancer (GVC) effects against residual disease. Among novel therapeutics for GVHD, oncolytic viruses such as myxoma virus (MYXV) are receiving increased attention due to their dual role in controlling GVHD while preserving or augmenting GVC. This review focuses on the molecular basis of GVHD, as well as state-of-the-art advances in developing novel therapies to prevent or control GVHD while minimizing impact on GVC. Recent literature regarding conventional and the emerging therapies are summarized, with special emphasis on virotherapy to prevent GVHD. Recent advances using preclinical models with oncolytic viruses such as MYXV to ameliorate the deleterious consequences of GVHD, while maintaining or improving the anti-cancer benefits of GVC will be reviewed.
Collapse
Affiliation(s)
- Nancy Y Villa
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Masmudur M Rahman
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Christopher R Cogle
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
233
|
Edinger M. Driving allotolerance: CAR-expressing Tregs for tolerance induction in organ and stem cell transplantation. J Clin Invest 2016; 126:1248-50. [PMID: 26999608 DOI: 10.1172/jci86827] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Regulatory T cells (Tregs) modulate the function of a variety of immune cells and are critical for maintaining self-tolerance and preventing the development of autoimmune disease. Due to their ability to suppress effector T cells, Tregs have been increasingly explored for clinical use to suppress alloresponses. While this approach has been promising in preclinical models and early clinical trials, widespread clinical use of Tregs has been limited by the low number of these cells in the periphery and the unknown frequency of allo-responsive Tregs. In this issue of the JCI, MacDonald and colleagues transduced human Tregs with a chimeric antigen receptor (CAR) that targets the HLA class I molecule A2. These CAR-expressing T cells were readily activated via CAR stimulation and exerted potent immunosuppressive effects when stimulated in vitro. In a murine model of hematopoietic stem cell transplantation, CAR-modified Tregs were more effective in preventing the development of graft-versus-host disease compared with polyclonal Tregs. The results of this study lay the groundwork for the further evaluation of CAR-expressing Tregs in the prevention or treatment of transplant complications.
Collapse
|
234
|
Preclinical models of acute and chronic graft-versus-host disease: how predictive are they for a successful clinical translation? Blood 2016; 127:3117-26. [PMID: 26994149 DOI: 10.1182/blood-2016-02-699082] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Abstract
Despite major advances in recent years, graft-versus-host disease (GVHD) remains a major life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT). To improve our therapeutic armory against GVHD, preclinical evidence is most frequently generated in mouse and large animal models of GVHD. However, because every model has shortcomings, it is important to understand how predictive the different models are and why certain findings in these models could not be translated into the clinic. Weaknesses of the animal GVHD models include the irradiation only-based conditioning regimen, the homogenous donor/recipient genetics in mice, canine or non-human primates (NHP), anatomic site of T cells used for transfer in mice, the homogenous microbial environment in mice housed under specific pathogen-free conditions, and the lack of pharmacologic GVHD prevention in control groups. Despite these major differences toward clinical allo-HCT, findings generated in animal models of GVHD have led to the current gold standards for GVHD prophylaxis and therapy. The homogenous nature of the preclinical models allows for reproducibility, which is key for the characterization of the role of a new cytokine, chemokine, transcription factor, microRNA, kinase, or immune cell population in the context of GVHD. Therefore, when carefully balancing reasons to apply small and large animal models, it becomes evident that they are valuable tools to generate preclinical hypotheses, which then have to be rigorously evaluated in the clinical setting. In this study, we discuss several clinical approaches that were motivated by preclinical evidence, novel NHP models and their advantages, and highlight the recent advances in understanding the pathophysiology of GVHD.
Collapse
|
235
|
Thompson PA, Rezvani K, Hosing CM, Oran B, Olson AL, Popat UR, Alousi AM, Shah ND, Parmar S, Bollard C, Hanley P, Kebriaei P, Cooper L, Kellner J, McNiece IK, Shpall EJ. Umbilical cord blood graft engineering: challenges and opportunities. Bone Marrow Transplant 2016; 50 Suppl 2:S55-62. [PMID: 26039209 DOI: 10.1038/bmt.2015.97] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We are entering a very exciting era in umbilical cord blood transplantation (UCBT), where many of the associated formidable challenges may become treatable by ex vivo graft manipulation and/or adoptive immunotherapy utilizing specific cellular products. We envisage the use of double UCBT rather than single UCBT for most patients; this allows for greater ability to treat larger patients as well as to manipulate the graft. Ex vivo expansion and/or fucosylation of one cord will achieve more rapid engraftment, minimize the period of neutropenia and also give certainty that the other cord will provide long-term engraftment/immune reconstitution. The non-expanded (and future dominant) cord could be chosen for characteristics such as better HLA matching to minimize GvHD, or larger cell counts to enable part of the unit to be utilized for the development of specific cellular therapies such as the production of virus-specific T-cells or chimeric-antigen receptor T-cells which are reviewed in this study.
Collapse
Affiliation(s)
- P A Thompson
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - K Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - C M Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - B Oran
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - A L Olson
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - U R Popat
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - A M Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - N D Shah
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - S Parmar
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - C Bollard
- Center for Cell Therapy and Department of Immunology, Baylor College of Medicine, Houston, TX, USA
| | - P Hanley
- Center for Cell Therapy and Department of Immunology, Baylor College of Medicine, Houston, TX, USA
| | - P Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - L Cooper
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - J Kellner
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - I K McNiece
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| | - E J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
236
|
Martelli MF, Ianni MD, Ruggeri L, Falzetti F, Carotti A, Reisner Y, Velardi A. Next generation HLA-haploidentical HSCT. Bone Marrow Transplant 2016; 50 Suppl 2:S63-6. [PMID: 26039211 DOI: 10.1038/bmt.2015.98] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Relapse is still the major cause of failure of allogeneic stem cell transplantation in high-risk acute leukemia patients. Indeed, whoever the donor and whatever the transplantation strategy, post-transplant relapse rates are ~30%, which is hardly satisfactory. The present phase 2 study analyzed the impact of adoptive immunotherapy with naturally occurring FoxP3+ T-regulatory cells (2 × 10(6) per kg) and conventional T lymphocytes (1 × 10(6) per kg) on prevention of GvHD and leukemia relapse in 43 high-risk adults undergoing full-haplotype mismatched transplantation without any post-transplant immunosuppression. Ninety-five percent of patients achieved full-donor type engraftment. Only 6/41 patients (15%) developed ⩾ grade II acute GvHD. Specific CD4(+) and CD8(+) for opportunistic pathogens emerged significantly earlier than after standard T-cell-depleted haplo-transplantation. The probability of disease-free survival was 0.56. At a median follow-up of 46 months (range 18-65 months), only 2/41 evaluable patients have relapsed. The cumulative incidence of relapse was significantly lower than in historical controls (0.05 vs 0.21; P = 0.03). These results demonstrate that the immunosuppressive potential of Tregs can be used to suppress GvHD without loss of the benefits of GvL activity. Humanized murine models provided insights into the mechanisms underlying separation of GvL from GvHD.
Collapse
Affiliation(s)
- M F Martelli
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - M D Ianni
- 1] Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy [2] Hematology Section, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - L Ruggeri
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - F Falzetti
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - A Carotti
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Y Reisner
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - A Velardi
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
237
|
Apostolova P, Zeiser R. The role of danger signals and ectonucleotidases in acute graft-versus-host disease. Hum Immunol 2016; 77:1037-1047. [PMID: 26902992 DOI: 10.1016/j.humimm.2016.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/09/2016] [Accepted: 02/18/2016] [Indexed: 12/28/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) represents the only curative treatment approach for many patients with benign or malignant diseases of the hematopoietic system. However, post-transplant morbidity and mortality are significantly increased by the development of acute graft-versus-host disease (GvHD). While alloreactive T cells act as the main cellular mediator of the GvH reaction, recent evidence suggests a critical role of the innate immune system in the early stages of GvHD initiation. Danger-associated molecular patterns released from the intracellular space as well as from the extracellular matrix activate antigen-presenting cells and set pro-inflammatory pathways in motion. This review gives an overview about danger signals representing therapeutic targets with a clinical perspective with a particular focus on extracellular nucleotides and ectonucleotidases.
Collapse
Affiliation(s)
- Petya Apostolova
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany.
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany.
| |
Collapse
|
238
|
Chen SY, Hsu WT, Chen YL, Chien CH, Chiang BL. Lymphocyte-activation gene 3(+) (LAG3(+)) forkhead box protein 3(-) (FOXP3(-)) regulatory T cells induced by B cells alleviates joint inflammation in collagen-induced arthritis. J Autoimmun 2016; 68:75-85. [PMID: 26908164 DOI: 10.1016/j.jaut.2016.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 02/01/2016] [Accepted: 02/01/2016] [Indexed: 11/18/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease in which dysregulated immune cells primarily target synovial joints. Despite recent advances in the treatment of RA, including the introduction of biologic therapies and employment of combination disease-modifying antirheumatic drug strategies, remission rates remain suboptimal. Previous studies have demonstrated that the adoptive transfer of induced regulatory T cells (iTregs) was effective in treating a murine model of collagen-induced arthritis (CIA). The objective of this study was to develop optimal potential iTreg-based therapy for CIA by adoptively transferring LAG3(+) Treg-of-B cells. B-cell-induced Treg-of-B cells expressed LAG3 but not Foxp3 (designated LAG3(+) Treg-of-B), and secreted IL-4, IL-10, and TGF-β. Furthermore, LAG3(+) Treg-of-B cells suppressed the proliferation of CD4(+)CD25(-) responder T cells through both LAG3 and IL-10 production. In the murine CIA model, adoptive transfer of LAG3(+) Treg-of-B cells alleviated the joint severity as well as local and systemic inflammation. Treatment with LAG3(+) Treg-of-B cells also promoted IL-10 production in lymphocytes isolated from the spleen and draining lymph nodes. Moreover, mice receiving LAG3(+) Treg-of-B cell treatment showed significantly less pronounced osteolysis in the hind footpads, which correlated with the downregulation of tartrate-resistant acid phosphatase expression. In conclusion, we identified a novel subset of Tregs for CIA treatment. This insight may facilitate exploring novel regulatory T-cell-based therapies for human autoimmune diseases.
Collapse
Affiliation(s)
- Szu-Ying Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Tseng Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Lien Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Hui Chien
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bor-Luen Chiang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
239
|
Grygorowicz MA, Biernacka M, Bujko M, Nowak E, Rymkiewicz G, Paszkiewicz-Kozik E, Borycka IS, Bystydzienski Z, Walewski J, Markowicz S. Human regulatory T cells suppress proliferation of B lymphoma cells. Leuk Lymphoma 2016; 57:1903-20. [DOI: 10.3109/10428194.2015.1121260] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
240
|
NK Cell and CD4+FoxP3+ Regulatory T Cell Based Therapies for Hematopoietic Stem Cell Engraftment. Stem Cells Int 2016; 2016:9025835. [PMID: 26880996 PMCID: PMC4736409 DOI: 10.1155/2016/9025835] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 10/22/2015] [Indexed: 11/18/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is a powerful therapy to treat multiple hematological diseases. The intensive conditioning regimens used to allow for donor hematopoietic stem cell (HSC) engraftment are often associated with severe toxicity, delayed immune reconstitution, life-threatening infections, and thus higher relapse rates. Additionally, due to the high incidence of graft versus host disease (GvHD), HCT protocols have evolved to prevent such disease that has a detrimental impact on antitumor and antiviral responses. Here, we analyzed the role of host T and natural killer (NK) cells in the rejection of donor HSC engraftment as well as the impact of donor regulatory T cells (Treg) and NK cells on HSC engraftment. We review some of the current strategies that utilize NK or Treg to improve allogeneic HCT therapy in order to accomplish better HSC engraftment and immune reconstitution and achieve a lower incidence of cancer relapse, opportunistic infections, and GvHD.
Collapse
|
241
|
Autoimmune Hepatitis: Progress from Global Immunosuppression to Personalised Regulatory T Cell Therapy. Can J Gastroenterol Hepatol 2016; 2016:7181685. [PMID: 27446862 PMCID: PMC4904688 DOI: 10.1155/2016/7181685] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 11/20/2015] [Indexed: 12/23/2022] Open
Abstract
Autoimmune hepatitis (AIH) is an immune mediated liver injury. The precise aetiology of AIH is still unknown but current evidence suggests both genetic and environmental factors are involved. Breakdown in peripheral self-tolerance, and impaired functions of FOXP3(+) regulatory T cell along with effector cell resistance to suppression at the tissue level seem to play an important role in AIH immunopathogenesis. AIH is predominantly a T lymphocytes driven disease but B lymphocytes are also involved in the immunopathology. Innate immune cells are crucial in the initial onset of disease and their response is followed by adaptive T (Th1, Th17, and cytotoxic T cells) and B cell responses evidenced by liver histology and peripheral blood serology. Standard treatment regimens involving steroid and immunosuppressive medications lead to global immune suppression requiring life-long therapy with many side effects. Biologic therapies have been attempted but duration of remission is short-lived. Future direction of diagnosis and treatment for AIH should be guided by "omics" and the immunology profile of the individual patient and clinicians should aim to deliver personalised medicine for their patients. Cell therapy such as infusion of autologous, antigen-specific, and liver-homing regulatory T cells to restore hepatic immune tolerance may soon be a potential future treatment for AIH patients.
Collapse
|
242
|
Swann JW, Garden OA. Novel immunotherapies for immune-mediated haemolytic anaemia in dogs and people. Vet J 2016; 207:13-9. [DOI: 10.1016/j.tvjl.2015.10.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
243
|
Dijke IE, Hoeppli RE, Ellis T, Pearcey J, Huang Q, McMurchy AN, Boer K, Peeters AMA, Aubert G, Larsen I, Ross DB, Rebeyka I, Campbell A, Baan CC, Levings MK, West LJ. Discarded Human Thymus Is a Novel Source of Stable and Long-Lived Therapeutic Regulatory T Cells. Am J Transplant 2016; 16:58-71. [PMID: 26414799 DOI: 10.1111/ajt.13456] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 06/19/2015] [Accepted: 07/14/2015] [Indexed: 01/25/2023]
Abstract
Regulatory T cell (Treg)-based therapy is a promising approach to treat many immune-mediated disorders such as autoimmune diseases, organ transplant rejection, and graft-versus-host disease (GVHD). Challenges to successful clinical implementation of adoptive Treg therapy include difficulties isolating homogeneous cell populations and developing expansion protocols that result in adequate numbers of cells that remain stable, even under inflammatory conditions. We investigated the potential of discarded human thymuses, routinely removed during pediatric cardiac surgery, to be used as a novel source of therapeutic Tregs. Here, we show that large numbers of FOXP3(+) Tregs can be isolated and expanded from a single thymus. Expanded thymic Tregs had stable FOXP3 expression and long telomeres, and suppressed proliferation and cytokine production of activated allogeneic T cells in vitro. Moreover, expanded thymic Tregs delayed development of xenogeneic GVHD in vivo more effectively than expanded Tregs isolated based on CD25 expression from peripheral blood. Importantly, in contrast to expanded blood Tregs, expanded thymic Tregs remained stable under inflammatory conditions. Our results demonstrate that discarded pediatric thymuses are an excellent source of therapeutic Tregs, having the potential to overcome limitations currently hindering the use of Tregs derived from peripheral or cord blood.
Collapse
Affiliation(s)
- I E Dijke
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada
| | - R E Hoeppli
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - T Ellis
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada
| | - J Pearcey
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada
| | - Q Huang
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - A N McMurchy
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - K Boer
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, the Netherlands
| | - A M A Peeters
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, the Netherlands
| | - G Aubert
- Terry Fox Laboratory, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - I Larsen
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada
| | - D B Ross
- Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - I Rebeyka
- Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - A Campbell
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - C C Baan
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, the Netherlands
| | - M K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - L J West
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
244
|
Pérol L, Piaggio E. New Molecular and Cellular Mechanisms of Tolerance: Tolerogenic Actions of IL-2. Methods Mol Biol 2016; 1371:11-28. [PMID: 26530792 DOI: 10.1007/978-1-4939-3139-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Interleukin-2 (IL-2) is an old molecule with brand new functions. Indeed, IL-2 has been first described as a T-cell growth factor but recent data pointed out that its main function in vivo is the maintenance of immune tolerance. Mechanistically, IL-2 is essential for the development and function of CD4(+) Foxp3(+) regulatory T cells (Treg cells) that are essential players in the control of immune responded to self, tumors, microbes and grafts. Treg cells are exquisitely sensitive to IL-2 due to their constitutive expression of the high affinity IL-2 receptor (IL-2R) and the new paradigm suggests that low-doses of IL-2 could selectively boost Treg cells in vivo. Consequently, a growing body of clinical research is aiming at using IL-2 at low doses as a tolerogenic drug to boost endogenous Treg cells in patients suffering from autoimmune or inflammatory conditions. In this manuscript, we briefly review IL-2/IL-2R biology and the role of IL-2 in the development, maintenance, and function of Treg cells; and also its effects on other immune cell populations such as CD4(+) T helper cells and CD8(+) memory T cells. Then, focusing on type 1 diabetes, we review the preclinical studies and clinical trials supporting the use of low-doses IL-2 as a tolerogenic immunotherapy. Finally, we discuss the limitations and future directions for IL-2 based immunotherapy.
Collapse
Affiliation(s)
- Louis Pérol
- INSERM U932, 26 rue d'Ulm, 75005, Paris, France.
- Institut Curie, Section Recherche, 26 rue d'Ulm, 75005, Paris, France.
| | - Eliane Piaggio
- INSERM U932, 26 rue d'Ulm, 75005, Paris, France
- Institut Curie, Section Recherche, 26 rue d'Ulm, 75005, Paris, France
| |
Collapse
|
245
|
Patel P, Mahmud D, Park Y, Yoshinaga K, Mahmud N, Rondelli D. Clinical grade isolation of regulatory T cells from G-CSF mobilized peripheral blood improves with initial depletion of monocytes. AMERICAN JOURNAL OF BLOOD RESEARCH 2015; 5:79-85. [PMID: 27069755 PMCID: PMC4769349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 10/27/2015] [Indexed: 06/05/2023]
Abstract
Clinical isolation of circulating CD4(+)CD25(+) regulatory T cells (Tregs) from peripheral blood mononuclear cells is usually performed by CD4(+) cell negative selection followed by CD25(+) cell positive selection. Although G-CSF mobilized peripheral blood (G-PBSC) contains a high number of Tregs, a high number of monocytes in G-PBSC limits Treg isolation. Using a small scale device (MidiMACS, Miltenyi) we initially demonstrated that an initial depletion of monocytes would be necessary to obtaina separation of CD4(+)CD25(+)FoxP3(+)CD127(-) cells from G-PBSC (G-Tregs) with a consistent purity >70% and inhibitory activity of T cell alloreactivity in-vitro. We then validated the same approach in a clinical scale setting by separating G-Tregs with clinically available antibodies to perform a CD8(+)CD19(+)CD14(+) cell depletion followed by CD25(+) cell selection (2-step process) or by adding an initial CD14(+) cell depletion (3-step process) using a CliniMACS column. The 3-step approach resulted in a better purity (81±12% vs. 35±33%) and yield (66% vs. 39%). Clinically isolated G-Tregs were also FoxP3(+)CD127(dim) and functionally suppressive in-vitro. Our findings suggest that a better and more consistent purity of Tregs can be achieved from G-PBSC by an initial single depletion of monocytes prior to selection of CD4(+)CD25(+) cells.
Collapse
Affiliation(s)
- Pritesh Patel
- Division of Hematology/Oncology, University of Illinois at ChicagoChicago, IL, USA
- Cancer Center, University of IllinoisChicago, IL, USA
| | - Dolores Mahmud
- Division of Hematology/Oncology, University of Illinois at ChicagoChicago, IL, USA
| | - Youngmin Park
- Hospital Stem Cell Laboratory, University of Illinois Hospital and Health Sciences SystemChicago, IL, USA
| | - Kazumi Yoshinaga
- Hospital Stem Cell Laboratory, University of Illinois Hospital and Health Sciences SystemChicago, IL, USA
| | - Nadim Mahmud
- Division of Hematology/Oncology, University of Illinois at ChicagoChicago, IL, USA
- Cancer Center, University of IllinoisChicago, IL, USA
- Hospital Stem Cell Laboratory, University of Illinois Hospital and Health Sciences SystemChicago, IL, USA
| | - Damiano Rondelli
- Division of Hematology/Oncology, University of Illinois at ChicagoChicago, IL, USA
- Cancer Center, University of IllinoisChicago, IL, USA
| |
Collapse
|
246
|
Inoue Y, Fuji S, Tanosaki R, Fukuda T. Pretransplant mogamulizumab against ATLL might increase the risk of acute GVHD and non-relapse mortality. Bone Marrow Transplant 2015; 51:725-7. [DOI: 10.1038/bmt.2015.315] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
247
|
Xu YJ, Li L, Chen Y, Fu B, Wu DS, Li XL, Zhao XL, Chen FP. Role of HMGB1 in regulation of STAT3 expression in CD4 + T cells from patients with aGVHD after allogeneic hematopoietic stem cell transplantation. Clin Immunol 2015; 161:278-83. [DOI: 10.1016/j.clim.2015.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 12/27/2022]
|
248
|
Inoue T, Ikegame K, Kaida K, Okada M, Yoshihara S, Tamaki H, Fujimori Y, Soma T, Ogawa H. Host Foxp3+CD4+ Regulatory T Cells Act as a Negative Regulator of Dendritic Cells in the Peritransplantation Period. THE JOURNAL OF IMMUNOLOGY 2015; 196:469-83. [PMID: 26621858 DOI: 10.4049/jimmunol.1402950] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 10/26/2015] [Indexed: 12/31/2022]
Abstract
Host Foxp3+CD4+ regulatory T cells (Tregs) have been shown to suppress graft-versus-host disease (GVHD) in experimental bone marrow transplantation (BMT) models; however, the detailed mechanism is unknown. To address this issue, we established a murine MHC-haploidentical BMT model (BDF1 (H-2b/d) → B6C3F1 (H-2b/k)), in which transplantation following conditioning with high-dose (13 Gy) or low-dose (5 Gy) total body irradiation corresponds to myeloablative stem cell transplantation (MAST) or reduced-intensity stem cell transplantation (RIST) BMT. All MAST recipients died of GVHD within 70 d, whereas RIST recipients developed almost no GVHD and survived for at least 3 mo. In this BMT model, we investigated the kinetics of immune cells in the mesenteric lymph nodes because GVHD was most prominent in the intestines. Host Tregs that survived after total body irradiation could proliferate transiently by day 4. Comparing the kinetics of immune cells among MAST, RIST, and anti-CD25 mAb-treated RIST, we found that the transiently surviving host Tregs were fully functional, closely contacted with host dendritic cells (DCs), and significantly restrained the maturation (CD80 and CD86 expression) of DCs in a dose-dependent manner. There was a positive correlation between the ratio of DCs to host Tregs and the extent of maturation of DCs. Host Tregs suppressed alloresponse mainly by contact inhibition. Host Tregs are already active in lymph nodes before transplantation and restrain the maturation of host DCs, thereby dampening the ability of DCs to activate allogeneic donor T cells and consequently reducing the magnitude of graft-versus-host reaction. Thus, host Tregs are negative regulators of host DCs that act in the peritransplantation period.
Collapse
Affiliation(s)
- Takayuki Inoue
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan; and
| | - Kazuhiro Ikegame
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan; and
| | - Katsuji Kaida
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan; and
| | - Masaya Okada
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan; and
| | - Satoshi Yoshihara
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan; and
| | - Hiroya Tamaki
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan; and
| | - Yoshihiro Fujimori
- Department of Transfusion Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Toshihiro Soma
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan; and
| | - Hiroyasu Ogawa
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo 663-8501, Japan; and
| |
Collapse
|
249
|
Haji S, Kiyasu J, Choi I, Suehiro Y, Toyoda K, Tsuda M, Takamatsu A, Nakashima Y, Miyoshi H, Shiratsuchi M, Yamasaki S, Uike N, Abe Y. Administration of an anti-CC chemokine receptor 4 monoclonal antibody, mogamulizumab, before allogeneic bone marrow transplantation for adult T-cell leukemia/lymphoma. Bone Marrow Transplant 2015; 51:432-4. [PMID: 26524267 DOI: 10.1038/bmt.2015.254] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- S Haji
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - J Kiyasu
- Department of Pathology, School of Medicine, Kurume University, Fukuoka, Japan.,Department of Hematology, Iizuka Hospital, Fukuoka, Japan
| | - I Choi
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - Y Suehiro
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - K Toyoda
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - M Tsuda
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - A Takamatsu
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - Y Nakashima
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - H Miyoshi
- Department of Pathology, School of Medicine, Kurume University, Fukuoka, Japan
| | - M Shiratsuchi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - S Yamasaki
- Department of Hematology, National Kyushu Medical Center, National Hospital Organization, Fukuoka, Japan
| | - N Uike
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - Y Abe
- Department of Hematology, National Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| |
Collapse
|
250
|
Noriega V, Martínez-Laperche C, Buces E, Pion M, Sánchez-Hernández N, Martín-Antonio B, Guillem V, Bosch-Vizcaya A, Bento L, González-Rivera M, Balsalobre P, Kwon M, Serrano D, Gayoso J, de la Cámara R, Brunet S, Rojas-Contreras R, Nieto JB, Martínez C, Gónzalez M, Espigado I, Vallejo JC, Sampol A, Jiménez-Velasco A, Urbano-Ispizua A, Solano C, Gallardo D, Díez-Martín JL, Buño I. The Genotype of the Donor for the (GT)n Polymorphism in the Promoter/Enhancer of FOXP3 Is Associated with the Development of Severe Acute GVHD but Does Not Affect the GVL Effect after Myeloablative HLA-Identical Allogeneic Stem Cell Transplantation. PLoS One 2015; 10:e0140454. [PMID: 26473355 PMCID: PMC4608671 DOI: 10.1371/journal.pone.0140454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/25/2015] [Indexed: 11/18/2022] Open
Abstract
The FOXP3 gene encodes for a protein (Foxp3) involved in the development and functional activity of regulatory T cells (CD4+/CD25+/Foxp3+), which exert regulatory and suppressive roles over the immune system. After allogeneic stem cell transplantation, regulatory T cells are known to mitigate graft versus host disease while probably maintaining a graft versus leukemia effect. Short alleles (≤(GT)15) for the (GT)n polymorphism in the promoter/enhancer of FOXP3 are associated with a higher expression of FOXP3, and hypothetically with an increase of regulatory T cell activity. This polymorphism has been related to the development of auto- or alloimmune conditions including type 1 diabetes or graft rejection in renal transplant recipients. However, its impact in the allo-transplant setting has not been analyzed. In the present study, which includes 252 myeloablative HLA-identical allo-transplants, multivariate analysis revealed a lower incidence of grade III-IV acute graft versus host disease (GVHD) in patients transplanted from donors harboring short alleles (OR = 0.26, CI 0.08-0.82, p = 0.021); without affecting chronic GVHD or graft versus leukemia effect, since cumulative incidence of relapse, event free survival and overall survival rates are similar in both groups of patients.
Collapse
Affiliation(s)
- Víctor Noriega
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Carolina Martínez-Laperche
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Elena Buces
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marjorie Pion
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Inmunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Beatriz Martín-Antonio
- Department of Hematology, Hospital Clinic, University of Barcelona, IDIBAPS, Instituto de Investigación Josep Carreras (IJC), Barcelona, Spain
| | - Vicent Guillem
- Department of Hematology and Medical Oncology, Hospital Clínico Universitario de Valencia, Universitat de Valencia, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Anna Bosch-Vizcaya
- Department of Hematology, ICO Girona, Hospital Josep Trueta, IDIBGI Foundation, Girona, Spain
| | - Leyre Bento
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Milagros González-Rivera
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- DNA Sequencing Core Facility, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Pascual Balsalobre
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Mi Kwon
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - David Serrano
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Jorge Gayoso
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | | | - Salut Brunet
- Department of Clinical Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - José B. Nieto
- Department of Hematology, Hospital Morales Meseguer, Murcia, Spain
| | | | - Marcos Gónzalez
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Ildefonso Espigado
- Department of Hematology and Hemotherapy, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Juan C. Vallejo
- Department of Hematology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Antonia Sampol
- Department of Hematology, Hospital Universitario Son Espases, Palma de Mallorca, Islas Baleares, Spain
| | | | - Alvaro Urbano-Ispizua
- Department of Hematology, Hospital Clinic, University of Barcelona, IDIBAPS, Instituto de Investigación Josep Carreras (IJC), Barcelona, Spain
| | - Carlos Solano
- Department of Hematology and Medical Oncology, Hospital Clínico Universitario de Valencia, Universitat de Valencia, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - David Gallardo
- Department of Hematology, ICO Girona, Hospital Josep Trueta, IDIBGI Foundation, Girona, Spain
| | - José L. Díez-Martín
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Ismael Buño
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | | |
Collapse
|