1
|
Çelik S, Kaynar L. Utilization of Clinical Data and Evaluation of Biomarkers in the Investigation of Graft-Versus-Host Disease Outcomes. Methods Mol Biol 2025; 2907:71-83. [PMID: 40100593 DOI: 10.1007/978-1-0716-4430-0_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Graft-versus-host disease (GVHD) is one of the most important obstacles after allogeneic hematopoietic stem cell transplantation (allo-HCT). The mortality rate is around 50%, especially in severe GVHD. One of the most important clinical outcomes in GVHD is non-relapse mortality (NRM). NRM was defined as death without evidence of relapse or progression. Kaplan Meier, log-rank test, and Cox model are used in survival analysis methods. There are various biomarkers that assess clinical outcomes of GVHD. Damage-associated molecular patterns, pathogen-associated molecular patterns, microRNAs, markers of endothelial dysfunction, cytokines, and their receptors are used to predict the occurrence of GVHD and clinical outcomes in GVHD. Furthermore, the utilization of panels that assess many biomarkers has proven to be successful in predicting the clinical outcomes of GVHD, particularly NRM.
Collapse
Affiliation(s)
- Serhat Çelik
- Department of Hematology, Yenimahalle Training and Research Hospital, Yıldırım Beyazıt University, Ankara, Turkey
| | - Leylagül Kaynar
- Department of Hematology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
2
|
Ehx G, Ritacco C, Baron F. Pathophysiology and preclinical relevance of experimental graft-versus-host disease in humanized mice. Biomark Res 2024; 12:139. [PMID: 39543777 PMCID: PMC11566168 DOI: 10.1186/s40364-024-00684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantations (allo-HCT) used for the treatment of hematological malignancies and other blood-related disorders. Until recently, the discovery of actionable molecular targets to treat GVHD and their preclinical testing was almost exclusively based on modeling allo-HCT in mice by transplanting bone marrow and splenocytes from donor mice into MHC-mismatched recipient animals. However, due to fundamental differences between human and mouse immunology, the translation of these molecular targets into the clinic can be limited. Therefore, humanized mouse models of GVHD were developed to circumvent this limitation. In these models, following the transplantation of human peripheral blood mononuclear cells (PBMCs) into immunodeficient mice, T cells recognize and attack mouse organs, inducing GVHD. Thereby, humanized mice provide a platform for the evaluation of the effects of candidate therapies on GVHD mediated by human immune cells in vivo. Understanding the pathophysiology of this xenogeneic GVHD is therefore crucial for the design and interpretation of experiments performed with this model. In this article, we comprehensively review the cellular and molecular mechanisms governing GVHD in the most commonly used model of xenogeneic GVHD: PBMC-engrafted NOD/LtSz-PrkdcscidIL2rγtm1Wjl (NSG) mice. By re-analyzing public sequencing data, we also show that the clonal expansion and the transcriptional program of T cells in humanized mice closely reflect those in humans. Finally, we highlight the strengths and limitations of this model, as well as arguments in favor of its biological relevance for studying T-cell reactions against healthy tissues or cancer cells.
Collapse
Affiliation(s)
- Grégory Ehx
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium.
| | - Caroline Ritacco
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium
| | - Frédéric Baron
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium
- Department of Medicine, Division of Hematology, CHU of Liege, University of Liege, Liege, Belgium
| |
Collapse
|
3
|
Koster EAS, von dem Borne PA, van Balen P, Marijt EWA, Tjon JML, Snijders TJF, van Lammeren D, Veelken H, Falkenburg JHF, Halkes CJM, de Wreede LC. Risk factors for graft-versus-host-disease after donor lymphocyte infusion following T-cell depleted allogeneic stem cell transplantation. Front Immunol 2024; 15:1335341. [PMID: 38545096 PMCID: PMC10966113 DOI: 10.3389/fimmu.2024.1335341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/13/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction Unmodified donor lymphocyte infusions (DLI) after allogeneic stem cell transplantation (alloSCT) can boost the beneficial Graft-versus-Leukemia (GvL) effect but may also induce severe Graft-versus-Host-Disease (GvHD). To improve the balance between GvL and GvHD, it is crucial to identify factors that influence the alloreactivity of DLI. Methods We investigated the effects of the presence of patient-derived antigen-presenting cells at time of DLI as estimated by the bone marrow (BM) chimerism status, lymphopenia as measured by the absolute lymphocyte count (ALC) at time of DLI, and the presence of a viral infection (de novo or reactivation) close to DLI on the risk of GvHD after DLI. The cohort consisted of patients with acute leukemia or myelodysplastic syndrome who prophylactically or pre-emptively received DLI as standard care after alemtuzumab-based alloSCT. In patients at high risk for relapse, DLI was administered at 3 months after alloSCT (n=88) with a dose of 0.3x106 or 0.15x106 T cells/kg in case of a related or unrelated donor, respectively. All other patients (n=76) received 3x106 or 1.5x106 T cells/kg, respectively, at 6 months after alloSCT. Results For both DLIs, patients with reduced-intensity conditioning and an unrelated donor had the highest risk of GvHD. For DLI given at three months, viral infection within 1 week before and 2 weeks after DLI was an additional significant risk factor (hazard ratio (HR) 3.66 compared to no viral infection) for GvHD. At six months after alloSCT, viral infections were rare and not associated with GvHD. In contrast, mixed BM chimerism (HR 3.63 for ≥5% mixed chimerism compared to full donor) was an important risk factor for GvHD after DLI given at six months after alloSCT. ALC of <1000x106/l showed a trend for association with GvHD after this DLI (HR 2.05 compared to ≥1000x106/l, 95% confidence interval 0.94-4.45). Furthermore, the data suggested that the presence of a viral infection close to the DLI at three months or ≥5% mixed chimerism at time of the DLI at six months correlated with the severity of GvHD, thereby increasing their negative impact on the current GvHD-relapse-free survival. Conclusion These data demonstrate that the risk factors for GvHD after DLI depend on the setting of the DLI.
Collapse
Affiliation(s)
- Eva A S Koster
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Peter van Balen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Erik W A Marijt
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Jennifer M L Tjon
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Hendrik Veelken
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Liesbeth C de Wreede
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
4
|
Kaulfuss M, Mietz J, Fabri A, Vom Berg J, Münz C, Chijioke O. The NK cell checkpoint NKG2A maintains expansion capacity of human NK cells. Sci Rep 2023; 13:10555. [PMID: 37386090 PMCID: PMC10310841 DOI: 10.1038/s41598-023-37779-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/27/2023] [Indexed: 07/01/2023] Open
Abstract
Human natural killer (NK) cells are cytotoxic effector cells that are increasingly harnessed in cancer immunotherapy. NKG2A/CD94 is an inhibitory receptor on NK cells that has established regulatory functions in the direct interaction with target cells when engaged with its ligand, the non-classical HLA class I molecule HLA-E. Here, we confirmed NKG2A as a checkpoint molecule in primary human NK cells and identified a novel role for NKG2A in maintaining NK cell expansion capacity by dampening both proliferative activity and excessive activation-induced cell death. Maintenance of NK cell expansion capacity might contribute to the preferential accumulation of human NKG2A+ NK cells after hematopoietic cell transplantation and enrichment of functionally impaired NK cells in human cancers. Functional silencing of NKG2A for cancer immunotherapy is highly attractive but will need to consider that this might also lead to a reduced survival by driving activation-induced cell death in targeted NK cells.
Collapse
Affiliation(s)
- Meike Kaulfuss
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Juliane Mietz
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Astrid Fabri
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
- Institute of Immunity & Transplantation, University College London Division of Infection & Immunity, London, UK
| | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zürich, Schlieren, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Obinna Chijioke
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland.
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
5
|
Haroun-Izquierdo A, Lanuza PM, Pfefferle A, Netskar H, Ask EH, Törlén J, Björklund A, Sohlberg E, Malmberg KJ. Effect of mTOR Inhibition with Sirolimus on Natural Killer Cell Reconstitution in Allogeneic Stem Cell Transplantation. Transplant Cell Ther 2023:S2666-6367(23)01201-0. [PMID: 36966873 DOI: 10.1016/j.jtct.2023.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 04/25/2023]
Abstract
Sirolimus is an inhibitor of the mammalian target of rapamycin (mTOR) and is emerging as a promising component of graft-versus-host disease (GVHD) prophylaxis regimens in the context of allogeneic hematopoietic stem cell transplantation (HSCT). Multiple studies have explored the clinical benefits of adding sirolimus to GVHD prophylaxis; however, detailed immunologic studies have not yet been carried out in this context. Mechanistically, mTOR is at the center of metabolic regulation in T cells and natural killer (NK) cells and is critical for their differentiation to mature effector cells. Therefore, close evaluation of the inhibition of mTOR in the context of immune reconstitution post-HSCT is warranted. In this work, we studied the effect of sirolimus on immune reconstitution using a biobank of longitudinal samples from patients receiving either tacrolimus/sirolimus (TAC/SIR) or cyclosporin A/methotrexate (CSA/MTX) as conventional GVHD prophylaxis. Healthy donor controls, donor graft material, and samples from 28 patients (14 with TAC/SIR, 14 with CSA/MTX) at 3 to 4 weeks and 34 to 39 weeks post- HSCT were collected. Multicolor flow cytometry was used to perform broad immune cell mapping, with a focus on NK cells. NK cell proliferation was evaluated over a 6-day in vitro homeostatic proliferation protocol. Furthermore, in vitro NK cell responses to cytokine stimulation or tumor cells were evaluated. Systems-level assessment of the immune repertoire revealed a deep and prolonged suppression (weeks 34 to 39 post-HSCT) of the naïve CD4 T cell compartment with relative sparing of regulatory T cells and enrichment of CD69+Ki-67+HLA-DR+ CD8 T cells, independent of the type of GVHD prophylaxis. Early after transplantation (weeks 3 to 4), while patients were still on TAC/SIR or CSA/MTX, we found a relative increase in less-differentiated CD56bright NK cells and NKG2A+CD57-KIR- CD56dim NK cells and a distinct loss of CD16 and DNAM-1 expression. Both regimens led to suppressed proliferative responses ex vivo and functional impairment with preferential loss of cytokine responsiveness and IFN-γ production. Patients who received TAC/SIR as GVHD prophylaxis showed delayed NK cell reconstitution with lower overall NK cell counts and fewer CD56bright and NKG2A+ CD56dim NK cells. Treatment with sirolimus- containing regimens generated similar immune cell profiles as conventional prophylaxis; however, the NK cell compartment seemed to be composed of slightly more mature NK cells. These effects were also present after the completion of GVHD prophylaxis, suggesting that mTOR inhibition with sirolimus leaves a lasting imprint on homeostatic proliferation and NK cell reconstitution following HSCT.
Collapse
Affiliation(s)
- Alvaro Haroun-Izquierdo
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Pilar M Lanuza
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Aline Pfefferle
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Herman Netskar
- Institute for Cancer Research, Department of Cancer Immunology, University of Oslo, Oslo University Hospital, Norway
| | - Eivind H Ask
- Institute for Cancer Research, Department of Cancer Immunology, University of Oslo, Oslo University Hospital, Norway
| | - Johan Törlén
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Andreas Björklund
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Ebba Sohlberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Karl-Johan Malmberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Institute for Cancer Research, Department of Cancer Immunology, University of Oslo, Oslo University Hospital, Norway.
| |
Collapse
|
6
|
Lee GH, Lee JY, Jang J, Kang YJ, Choi SA, Kim HC, Park S, Kim MS, Lee W. Anti‐thymocyte globulin‐mediated immunosenescent alterations of T cells in kidney transplant patients. Clin Transl Immunology 2022; 11:e1431. [PMCID: PMC9686013 DOI: 10.1002/cti2.1431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 10/07/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022] Open
Abstract
Objectives Kidney transplant (KT) is the most effective treatment for end‐stage renal disease. The immunosuppressant anti‐thymocyte globulin (ATG) has been applied for induction therapy to reduce the risk of acute transplant rejection for patients at high immunological risk. Despite its putative role in replicative stress during immune reconstitution, the effects of ATG on T‐cell immunosenescent changes remain to be understood. Methods Phenotypic and functional features of senescent T cells were examined by flow cytometry in 116 healthy controls (HC) and 95 KT patients for comparative analysis according to ATG treatment and CMV reactivation. The TCR repertoire was analysed in peripheral blood mononuclear cells (PBMCs) of KT patients. Results T cells of KT patients treated with ATG (ATG+) show typical immunosenescent features, accumulation of CD28−, CD85j+ or CD57+ T cells, and imbalance of functional T‐cell subsets, compared with untreated KT patients (ATG−). Plasma IL‐15 and CMV‐IgG levels were higher in KT patients than in HCs, and the IL‐15 level positively correlated with the frequency of CD28− T cells in KT patients. ATG+ patients had a higher prevalence of CMV reactivation, which is associated with an increased frequency of CD28− T cells. As a result, ATG+ patients had expanded CMV‐specific T cells and decreased TCR diversity. However, proliferation, cytokine‐producing capacity and polyfunctionality of T cells were preserved in ATG+ patients. Conclusion Our findings suggest that ATG treatment contributes to the accumulation of senescent T cells, which may have lifelong clinical implications in KT patients. Thus, these patients require long‐term and comprehensive immune monitoring.
Collapse
Affiliation(s)
- Ga Hye Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical SciencesSeoul National University College of MedicineSeoulSouth Korea,Department of Microbiology and ImmunologySeoul National University College of MedicineSeoulSouth Korea
| | - Jee Youn Lee
- Department of SurgeryKangbuk Samsung Hospital, Sungkyunkwan University School of MedicineSeoulSouth Korea
| | - Jiyeon Jang
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical SciencesSeoul National University College of MedicineSeoulSouth Korea,Department of Microbiology and ImmunologySeoul National University College of MedicineSeoulSouth Korea
| | - Yeon Jun Kang
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical SciencesSeoul National University College of MedicineSeoulSouth Korea,Department of Microbiology and ImmunologySeoul National University College of MedicineSeoulSouth Korea
| | - Seung Ah Choi
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical SciencesSeoul National University College of MedicineSeoulSouth Korea,Department of Microbiology and ImmunologySeoul National University College of MedicineSeoulSouth Korea
| | - Hyeon Chang Kim
- Department of Preventive MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Sungha Park
- Division of Cardiology, Severance Cardiovascular HospitalYonsei University Health SystemSeoulSouth Korea
| | - Myoung Soo Kim
- Department of SurgeryYonsei University College of MedicineSeoulSouth Korea
| | - Won‐Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical SciencesSeoul National University College of MedicineSeoulSouth Korea,Department of Microbiology and ImmunologySeoul National University College of MedicineSeoulSouth Korea,Cancer Research Institute, Ischemic/Hypoxic Disease Institute, and Institute of Infectious DiseasesSeoul National University College of Medicine; Seoul National University Hospital Biomedical Research InstituteSeoulSouth Korea
| |
Collapse
|
7
|
Characterization of Hepatic Dysfunction in Subjects Diagnosed With Chronic GVHD by NIH Consensus Criteria. Transplant Cell Ther 2022; 28:747.e1-747.e10. [DOI: 10.1016/j.jtct.2022.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 11/23/2022]
|
8
|
Paul B, Rodriguez C, Usmani SZ. BCMA-Targeted Biologic Therapies: The Next Standard of Care in Multiple Myeloma Therapy. Drugs 2022; 82:613-631. [PMID: 35412114 PMCID: PMC9554894 DOI: 10.1007/s40265-022-01697-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 11/03/2022]
Abstract
With recent advances in myeloma therapy, patients can achieve long-term remissions, but eventually relapses will occur. Triple-class refractory myeloma (disease that is refractory to an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody) and penta-refractory myeloma (disease that is refractory to two proteasome inhibitors, two immunomodulatory agents, and an anti-CD38 antibody) are associated with a particularly poor prognosis, and novel treatments are desperately needed for these patients. Targeting B cell maturation antigen (BCMA), which is ubiquitously expressed on plasma cells, has emerged as a well-tolerated and highly efficacious strategy in patients with relapsed and refractory myeloma. Several mechanisms of targeting BCMA are currently under investigation, including antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T cells and natural killer (NK) cells, all with unique side effect profiles. Early phase clinical trials showed unprecedented response rates in highly refractory myeloma patients, leading to the recent approvals of some of these agents. Still, many questions remain with regard to this target, including how best to target it, how to treat patients who have progressed on a BCMA-targeting therapy, and whether response rates will deepen if these agents are used in earlier lines of therapy. In this review, we examine the rationale for targeting BCMA and summarize the data for several agents across multiple classes of BCMA-targeting therapeutics, paying special attention to the diverse mechanisms and unique challenges of each therapeutic class.
Collapse
Affiliation(s)
- Barry Paul
- Division of Plasma Cell Disorders, Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | | | - Saad Z Usmani
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
9
|
Kielsen K, Oostenbrink LVE, von Asmuth EGJ, Jansen-Hoogendijk AM, van Ostaijen-Ten Dam MM, Ifversen M, Heilmann C, Schilham MW, van Halteren AGS, Bredius RGM, Lankester AC, Jol-van der Zijde CM, van Tol MJD, Müller K. IL-7 and IL-15 Levels Reflect the Degree of T Cell Depletion during Lymphopenia and Are Associated with an Expansion of Effector Memory T Cells after Pediatric Hematopoietic Stem Cell Transplantation. THE JOURNAL OF IMMUNOLOGY 2021; 206:2828-2838. [PMID: 34108260 DOI: 10.4049/jimmunol.2001077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/22/2021] [Indexed: 11/19/2022]
Abstract
Differentially and functionally distinct T cell subsets are involved in the development of complications after allogeneic hematopoietic stem cell transplantation (HSCT), but little is known about factors regulating their recovery after HSCT. In this study, we investigated associations between immune-regulating cytokines, T cell differentiation, and clinical outcomes. We included 80 children undergoing allogeneic HSCT for acute leukemia using bone marrow or peripheral blood stem cells grafted from a matched sibling or unrelated donor. Cytokines (IL-7, IL-15, IL-18, SCF, IL-6, IL-2, and TNF-α) and active anti-thymocyte globulin (ATG) levels were longitudinally measured along with extended T cell phenotyping. The cytokine profiles showed a temporary rise in IL-7 and IL-15 during lymphopenia, which was strongly dependent on exposure to active ATG. High levels of IL-7 and IL-15 from graft infusion to day +30 were predictive of slower T cell recovery during the first 2 mo post-HSCT; however, because of a major expansion of memory T cell stages, only naive T cells remained decreased after 3 mo (p < 0.05). No differential effect was seen on polarization of CD4+ T cells into Th1, Th2, or Th17 cells or regulatory T cells. Low levels of IL-7 and IL-15 at day +14 were associated with acute graft-versus-host disease grades II-IV in ATG-treated patients (p = 0.0004 and p = 0.0002, respectively). Children with IL-7 levels comparable to healthy controls at day +14 post-HSCT were less likely to develop EBV reactivation posttransplant. These findings suggest that quantification of IL-7 and IL-15 may be useful as biomarkers in assessing the overall T cell depletion and suggest a potential for predicting complications after HSCT.
Collapse
Affiliation(s)
- Katrine Kielsen
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; .,Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; and
| | - Lisa V E Oostenbrink
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Erik G J von Asmuth
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Anja M Jansen-Hoogendijk
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Monique M van Ostaijen-Ten Dam
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Marianne Ifversen
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Carsten Heilmann
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Marco W Schilham
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Astrid G S van Halteren
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Robbert G M Bredius
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Arjan C Lankester
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Cornelia M Jol-van der Zijde
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten J D van Tol
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Klaus Müller
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; and
| |
Collapse
|
10
|
Yakoub-Agha I, Chabannon C, Bader P, Basak GW, Bonig H, Ciceri F, Corbacioglu S, Duarte RF, Einsele H, Hudecek M, Kersten MJ, Köhl U, Kuball J, Mielke S, Mohty M, Murray J, Nagler A, Robinson S, Saccardi R, Sanchez-Guijo F, Snowden JA, Srour M, Styczynski J, Urbano-Ispizua A, Hayden PJ, Kröger N. Management of adults and children undergoing chimeric antigen receptor T-cell therapy: best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE). Haematologica 2020; 105:297-316. [PMID: 31753925 PMCID: PMC7012497 DOI: 10.3324/haematol.2019.229781] [Citation(s) in RCA: 242] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells are a novel class of anti-cancer therapy in which autologous or allogeneic T cells are engineered to express a CAR targeting a membrane antigen. In Europe, tisagenlecleucel (Kymriah™) is approved for the treatment of refractory/relapsed acute lymphoblastic leukemia in children and young adults as well as relapsed/refractory diffuse large B-cell lymphoma, while axicabtagene ciloleucel (Yescarta™) is approved for the treatment of relapsed/refractory high-grade B-cell lymphoma and primary mediastinal B-cell lymphoma. Both agents are genetically engineered autologous T cells targeting CD19. These practical recommendations, prepared under the auspices of the European Society of Blood and Marrow Transplantation, relate to patient care and supply chain management under the following headings: patient eligibility, screening laboratory tests and imaging and work-up prior to leukapheresis, how to perform leukapheresis, bridging therapy, lymphodepleting conditioning, product receipt and thawing, infusion of CAR T cells, short-term complications including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, antibiotic prophylaxis, medium-term complications including cytopenias and B-cell aplasia, nursing and psychological support for patients, long-term follow-up, post-authorization safety surveillance, and regulatory issues. These recommendations are not prescriptive and are intended as guidance in the use of this novel therapeutic class.
Collapse
Affiliation(s)
| | - Christian Chabannon
- Institut Paoli-Calmettes & Module Biothérapies, INSERM CBT-1409, Centre d'Investigations Cliniques de Marseille, Marseille, France
| | - Peter Bader
- Clinic for Children and Adolescents, University Children's Hospital, Frankfurt, Germany
| | - Grzegorz W Basak
- Department of Hematology, Oncology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology of Goethe University and German Red Cross Blood Service, Frankfurt, Germany
| | - Fabio Ciceri
- Università Vita-Salute San Raffaele, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Selim Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Regensburg, Regensburg, Germany
| | - Rafael F Duarte
- Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Hermann Einsele
- Medizinische Klinikund Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinikund Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Marie José Kersten
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam and LYMMCARE, Amsterdam, the Netherlands
| | - Ulrike Köhl
- Fraunhofer Institute for Cellular Therapeutics and Immunology (IZI) and Institute of Clinical Immunology, University of Leipzig, Leipzig as well as Institute for Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Jürgen Kuball
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stephan Mielke
- Department of Laboratory Medicine/Department of Cell Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Mohamad Mohty
- Hôpital Saint-Antoine, AP-HP, Sorbonne Université, INSERM UMRS 938, Paris, France
| | | | - Arnon Nagler
- The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | - Fermin Sanchez-Guijo
- IBSAL-Hospital Universitario de Salamanca, CIC, Universidad de Salamanca, Salamanca, Spain
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Micha Srour
- Service des Maladies du Sang, CHU de Lille, Lille, France
| | - Jan Styczynski
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University Torun, Bydgoszcz, Poland
| | | | - Patrick J Hayden
- Department. of Hematology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg, Hamburg, Germany
| |
Collapse
|
11
|
Zhao XS, Huang XJ. Seeking biomarkers for acute graft-versus-host disease: where we are and where we are heading? Biomark Res 2019; 7:17. [PMID: 31406575 PMCID: PMC6685226 DOI: 10.1186/s40364-019-0167-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) is one of the most important complications after allogeneic hematopoietic stem cell transplantation (allo-HSCT), which would seriously affect the clinical outcomes of patients. Early diagnosis and early intervention are keys for improving its curative efficacy. Thus, seeking the biomarkers of aGVHD that can accurately identify and diagnose aGVHD is very important to guiding the intervention and treatment of aGVHD. For the past decades, many studies have focused on searching for aGVHD-related biological markers to assist in diagnosis, early warning, and risk stratification. Unfortunately, until now, no reliable aGVHD biomarker is available that is recognized and widely used in clinical practice. With the continuous development of biological technology, as well as our in-depth understanding of the pathophysiologic mechanism of aGVHD, the selection, examination and application of biological markers have changed much. In this review, we summarized the progress of aGVHD biological marker screening, identification, preliminary clinical application, and look forward to a promising development direction in the future.
Collapse
Affiliation(s)
- Xiao-Su Zhao
- 1Peking University Peopl's Hospital, Peking University Institute of Hematology, No.11 Xizhimen South Street, Beijing, 100044 China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,3Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- 1Peking University Peopl's Hospital, Peking University Institute of Hematology, No.11 Xizhimen South Street, Beijing, 100044 China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,3Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,4Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
12
|
Phan TL, Carlin K, Ljungman P, Politikos I, Boussiotis V, Boeckh M, Shaffer ML, Zerr DM. Human Herpesvirus-6B Reactivation Is a Risk Factor for Grades II to IV Acute Graft-versus-Host Disease after Hematopoietic Stem Cell Transplantation: A Systematic Review and Meta-Analysis. Biol Blood Marrow Transplant 2018; 24:2324-2336. [PMID: 29684567 PMCID: PMC8934525 DOI: 10.1016/j.bbmt.2018.04.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022]
Abstract
Graft-versus-host disease (GVHD) is an important cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). Many studies have suggested that human herpesvirus-6B (HHV-6B) plays a role in acute GVHD (aGVHD) after HCT. Our objective was to systematically summarize and analyze evidence regarding HHV-6B reactivation and development of aGVHD. PubMed and EMBASE databases were searched using terms for HHV-6, HCT, and aGVHD, yielding 865 unique results. Case reports, reviews, articles focusing on inherited chromosomally integrated HHV-6, poster presentations, and articles not published in English were excluded. The remaining 467 articles were reviewed for the following requirements: a statistical analysis of HHV-6B reactivation and a GVHD was described, HHV-6B reactivation was defined by PCR, and blood (plasma, serum, or peripheral blood mononuclear cells) was used for HHV-6B PCR. Data were abstracted from publications that met these criteria (n = 33). Publications were assigned to 1 of 3 groups: (1) HHV-6B reactivation was analyzed as a time-dependent risk factor for subsequent aGVHD (n = 14), (2) aGVHD was analyzed as a time-dependent risk factor for subsequent HHV-6B reactivation (n = 1), and (3) analysis without temporal specification (n = 18). A statistically significant association (P < .05) between HHV-6B reactivation and aGVHD was observed in 10 of 14 studies (71%) in group 1, 0 of 1 study (0%) in Group 2, and 8 of 18 studies (44.4%) in Group 3. Of the 14 studies that analyzed HHV-6B as a risk factor for subsequent aGVHD, 11 performed a multivariate analysis and reported a hazard ratio, which reached statistical significance in 9 of these s tudies. Meta-analysis of these 11 studies demonstrated a statistically significant association between HHV-6B and subsequent grades II to IV aGVHD (hazard ratio, 2.65; 95% confidence interval, 1.89 to 3.72; P < .001).HHV-6B reactivation is associated with aGVHD, and when studies have a temporal component to their design, HHV-6B reactivation is associated with subsequent aGVHD. Further research is needed to investigate whether antiviral prophylaxis reduces incidence or severity of aGVHD.
Collapse
Affiliation(s)
- Tuan L Phan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana; HHV-6 Foundation, Santa Barbara, California
| | - Kristen Carlin
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, Washington
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden; Division of Hematology, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Politikos
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vicki Boussiotis
- Department of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Michael Boeckh
- Department of Medicine, Vaccine and Infectious Disease and Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Michele L Shaffer
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, Washington; Department of Statistics, University of Washington, Seattle, Washington
| | - Danielle M Zerr
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, Washington; Department of Pediatrics, University of Washington, Seattle, Washington.
| |
Collapse
|
13
|
He FC, Holtan SG. Biomarkers in Graft-Versus-Host Disease: from Prediction and Diagnosis to Insights into Complex Graft/Host Interactions. Curr Hematol Malig Rep 2018; 13:44-52. [DOI: 10.1007/s11899-018-0433-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
14
|
Ullrich E, Abendroth B, Rothamer J, Huber C, Büttner-Herold M, Buchele V, Vogler T, Longerich T, Zundler S, Völkl S, Beilhack A, Rose-John S, Wirtz S, Weber GF, Ghimire S, Kreutz M, Holler E, Mackensen A, Neurath MF, Hildner K. BATF-dependent IL-7RhiGM-CSF+ T cells control intestinal graft-versus-host disease. J Clin Invest 2018; 128:916-930. [PMID: 29376889 DOI: 10.1172/jci89242] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 12/12/2017] [Indexed: 11/17/2022] Open
Abstract
Acute graft-versus-host disease (GVHD) represents a severe, T cell-driven inflammatory complication following allogeneic hematopoietic cell transplantation (allo-HCT). GVHD often affects the intestine and is associated with a poor prognosis. Although frequently detectable, proinflammatory mechanisms exerted by intestinal tissue-infiltrating Th cell subsets remain to be fully elucidated. Here, we show that the Th17-defining transcription factor basic leucine zipper transcription factor ATF-like (BATF) was strongly regulated across human and mouse intestinal GVHD tissues. Studies in complete MHC-mismatched and minor histocompatibility-mismatched (miHA-mismatched) GVHD models revealed that BATF-expressing T cells were functionally indispensable for intestinal GVHD manifestation. Mechanistically, BATF controlled the formation of colon-infiltrating, IL-7 receptor-positive (IL-7R+), granulocyte-macrophage colony-stimulating factor-positive (GM-CSF+), donor T effector memory (Tem) cells. This T cell subset was sufficient to promote intestinal GVHD, while its occurrence was largely dependent on T cell-intrinsic BATF expression, required IL-7-IL-7R interaction, and was enhanced by GM-CSF. Thus, this study identifies BATF-dependent pathogenic GM-CSF+ effector T cells as critical promoters of intestinal inflammation in GVHD and hence putatively provides mechanistic insight into inflammatory processes previously assumed to be selectively Th17 driven.
Collapse
Affiliation(s)
- Evelyn Ullrich
- Department of Medicine 5, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany.,Children's Hospital, Department of Pediatric Stem Cell Transplantation and Immunology, and.,LOEWE Center for Cell and Gene Therapy, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Benjamin Abendroth
- Department of Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research, Erlangen, Germany
| | - Johanna Rothamer
- Department of Medicine 5, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany.,Children's Hospital, Department of Pediatric Stem Cell Transplantation and Immunology, and.,LOEWE Center for Cell and Gene Therapy, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Carina Huber
- Department of Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research, Erlangen, Germany
| | - Maike Büttner-Herold
- Institute of Pathology, Department of Nephropathology, University Hospital Erlangen, Erlangen, Germany
| | - Vera Buchele
- Department of Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research, Erlangen, Germany
| | - Tina Vogler
- Department of Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research, Erlangen, Germany
| | - Thomas Longerich
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research, Erlangen, Germany
| | - Simon Völkl
- Department of Medicine 5, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Beilhack
- Center for Interdisciplinary Clinical Research, Würzburg University, Würzburg, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Stefan Wirtz
- Department of Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research, Erlangen, Germany
| | - Georg F Weber
- Department of Surgery, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Sakhila Ghimire
- Department of Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Marina Kreutz
- Department of Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Ernst Holler
- Department of Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Mackensen
- Department of Medicine 5, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research, Erlangen, Germany
| | - Kai Hildner
- Department of Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research, Erlangen, Germany
| |
Collapse
|
15
|
Yakoub-Agha I, Ferrand C, Chalandon Y, Ballot C, Castilla Llorente C, Deschamps M, Gauthier J, Labalette M, Larghero J, Maheux C, Moreau AS, Varlet P, Pétillon MO, Pinturaud M, Rubio MT, Chabannon C. Prérequis nécessaires pour la mise en place de protocoles de recherche clinique évaluant des thérapies cellulaires et géniques par lymphocytes T dotés de récepteur chimérique à l’antigène (CAR T-cells) : recommandations de la Société francophone de greffe de moelle et de thérapie cellulaire (SFGM-TC). Bull Cancer 2017; 104:S43-S58. [DOI: 10.1016/j.bulcan.2017.10.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 11/30/2022]
|
16
|
Kielsen K, Shamim Z, Thiant S, Faucher S, Decker W, Christensen IJ, Ryder LP, Yakoub-Agha I, Müller K. Soluble Interleukin-7 receptor levels and risk of acute graft-versus-host disease after allogeneic haematopoietic stem cell transplantation. Clin Immunol 2017; 187:26-32. [PMID: 28863969 DOI: 10.1016/j.clim.2017.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 07/10/2017] [Accepted: 08/28/2017] [Indexed: 01/05/2023]
Abstract
Interleukin-7 is a cytokine essential for T cell homeostasis. IL-7 binds to cellular IL-7 receptors in competition with a soluble form of the receptor (sIL-7Rα). We hypothesized that altered sIL-7Rα levels may cause adverse outcomes in patients undergoing HSCT. In parallel, we investigated the impact of the IL-7Rα SNP rs6897932, which has been associated with release of IL-7R. The sIL-7Rα levels decreased during HSCT (from 114ng/ml before to 48ng/ml at day +14 (P<0.0001)). This pattern was inversely mirrored by IL-7. The IL-7/sIL-7Rα ratio at day +14 was significantly higher in patients developing grades II-IV aGVHD (OR=4.3, P=0.026). Furthermore, donor carriage of the rs6897932 T allele was associated with reduced sIL-7Rα levels, increased risk of grades II-IV aGVHD (OR=2.4, P=0.055) and increased transplant-related mortality (CC=4.5%, CT=21.4% and TT=27.3%, P=0.0037). In conclusion, this study suggests an impact of sIL-7Rα levels and rs6897932 donor genotype on alloreactivity and outcome after HSCT.
Collapse
Affiliation(s)
- Katrine Kielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark; Haematopoietic Cell Transplantation and Primary Immune Deficiency, Department of Paediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark.
| | - Zaiba Shamim
- Institute for Inflammation Research, Center for Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark; The Tissue Typing Laboratory, Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
| | - Stephanie Thiant
- Department of Microbiology and Immunology, Hôpital Maisonneuve-Rosemont, Boulevard de l'Assomption 5415, Montréal, Quebec, Canada
| | - Sylvie Faucher
- Biologics and Genetic Therapies Directorate, Health Canada, Address Locator 0900C2, Ottawa, Ontario, Canada
| | - Wendy Decker
- National HIV and Retrovirology Laboratories, Public Health Agency of Canada, Eglantine Drw 100, Tunney's Pasture, Ottawa, Ontario, Canada
| | - Ib Jarle Christensen
- Department of Surgical Gastroenterology, Hvidovre Hospital, Kettegård Allé 30, Hvidovre, Denmark
| | - Lars Peter Ryder
- The Tissue Typing Laboratory, Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
| | - Ibrahim Yakoub-Agha
- Laboratory of Immunology, Center of Biology and Pathology, Centre Hospitalier Régional Universitaire de Lille, Avenue Oscar Lambret 1, 59037 Lille Cedex, France
| | - Klaus Müller
- Institute for Inflammation Research, Center for Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark; Haematopoietic Cell Transplantation and Primary Immune Deficiency, Department of Paediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
| |
Collapse
|
17
|
Clinical and immunologic impact of CCR5 blockade in graft-versus-host disease prophylaxis. Blood 2017; 129:906-916. [PMID: 28057639 DOI: 10.1182/blood-2016-08-735076] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/20/2016] [Indexed: 01/10/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Lymphocyte trafficking via chemokine receptors such as CCR5 plays a critical role in alloreactive responses, and previous data suggest that CCR5 blockade with maraviroc results in a low incidence of visceral GVHD. However, the full scope of clinical and immunologic effects of CCR5 blockade in HSCT has not been described. We compared a cohort of patients enrolled on a trial of reduced-intensity allo-HSCT with standard GVHD prophylaxis plus maraviroc to a contemporary control cohort receiving standard GVHD prophylaxis alone. Maraviroc treatment was associated with a lower incidence of acute GVHD without increased risk of disease relapse, as well as reduced levels of gut-specific markers. At day 30, maraviroc treatment increased CCR5 expression on T cells and dampened T-cell activation in peripheral blood without impairing early immune reconstitution or increasing risk for infections. Patients who developed acute GVHD despite maraviroc prophylaxis showed increased T-cell activation, naive T-cell skewing, and elevated serum CXCL9 and CXCL10 levels. Collectively, these data suggest that maraviroc effectively protects against GVHD by modulating alloreactive donor T-cell responses, and that CXCR3 signaling may be an important resistance mechanism to CCR5 blockade in GVHD.
Collapse
|
18
|
The Role of Biomarkers in the Diagnosis and Risk Stratification of Acute Graft-versus-Host Disease: A Systematic Review. Biol Blood Marrow Transplant 2016; 22:1552-1564. [PMID: 27158050 DOI: 10.1016/j.bbmt.2016.04.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/26/2016] [Indexed: 12/20/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is an increasingly used curative modality for hematologic malignancies and other benign conditions. Attempts to reduce morbidity and mortality and improve survival in patients undergoing HCT are crucial. The ability to diagnose acute graft-versus-host disease (aGVHD) in a timely manner, or to even predict aGVHD before clinical manifestations, along with the accurate stratification of these patients, are critical steps to improve the treatment and outcomes of these patients. Many novel biomarkers that may help achieve these goals have been studied recently. This overview is intended to assist clinicians and investigators by providing a comprehensive review and analytical interpretation of the current knowledge concerning aGVHD and biomarkers likely to prove useful in diagnosis and risk stratification of this condition, along with the difficulties that hamper this approach.
Collapse
|
19
|
Thiant S, Moutuou MM, Leboeuf D, Guimond M. Homeostatic cytokines in immune reconstitution and graft-versus-host disease. Cytokine 2016; 82:24-32. [PMID: 26795458 DOI: 10.1016/j.cyto.2016.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/07/2016] [Accepted: 01/07/2016] [Indexed: 12/15/2022]
Abstract
For numerous patients, allogeneic stem cell transplantation (SCT) is the only therapeutic option that could potentially cure their disease. Despite significant progress made in clinical management of allogeneic SCT, acute graft-versus-host disease (aGVHD) remains the second cause of death after disease recurrence. aGVHD is highly immunosuppressive and the adverse effect of allogeneic SCT on T cell regeneration is typically more important than the levels of immunosuppression normally seen after autologous SCT. In these patients, immune reconstitution often takes several years to occur and restoring immunocompetence after allogeneic SCT represents an important challenge, principally because clinical options are limited and current methods used to accelerate immune reconstitution are associated with increased GVHD. Interleukin-7 and IL-15 are both under clinical investigation and demonstrate the greatest potential on peripheral T cells regeneration in mice and humans. However, awareness has been raised about the use of IL-7 and IL-15 after allogeneic SCT with regards to potential adverse effects on aGVHD. In this review, we will discuss about recent progress made in lymphocyte regeneration, the critical role played by IL-7 and IL-15 in T cell homeostasis and how these cytokines could be used to improve immune reconstitution after allogeneic SCT.
Collapse
Affiliation(s)
- Stéphanie Thiant
- Maisonneuve-Rosemont Research Center, Montreal, Quebec, Canada; Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Moutuaata M Moutuou
- Maisonneuve-Rosemont Research Center, Montreal, Quebec, Canada; Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Dominique Leboeuf
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Martin Guimond
- Maisonneuve-Rosemont Research Center, Montreal, Quebec, Canada; Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
20
|
Allgäuer A, Schreiner E, Ferrazzi F, Ekici AB, Gerbitz A, Mackensen A, Völkl S. IL-7 Abrogates the Immunosuppressive Function of Human Double-Negative T Cells by Activating Akt/mTOR Signaling. THE JOURNAL OF IMMUNOLOGY 2015; 195:3139-48. [DOI: 10.4049/jimmunol.1501389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/02/2015] [Indexed: 11/19/2022]
|
21
|
Zeiser R, Burchert A, Lengerke C, Verbeek M, Maas-Bauer K, Metzelder SK, Spoerl S, Ditschkowski M, Ecsedi M, Sockel K, Ayuk F, Ajib S, de Fontbrune FS, Na IK, Penter L, Holtick U, Wolf D, Schuler E, Meyer E, Apostolova P, Bertz H, Marks R, Lübbert M, Wäsch R, Scheid C, Stölzel F, Ordemann R, Bug G, Kobbe G, Negrin R, Brune M, Spyridonidis A, Schmitt-Gräff A, van der Velden W, Huls G, Mielke S, Grigoleit GU, Kuball J, Flynn R, Ihorst G, Du J, Blazar BR, Arnold R, Kröger N, Passweg J, Halter J, Socié G, Beelen D, Peschel C, Neubauer A, Finke J, Duyster J, von Bubnoff N. Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey. Leukemia 2015; 29:2062-8. [PMID: 26228813 DOI: 10.1038/leu.2015.212] [Citation(s) in RCA: 415] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 07/23/2015] [Accepted: 07/24/2015] [Indexed: 12/15/2022]
Abstract
Despite major improvements in allogeneic hematopoietic cell transplantation over the past decades, corticosteroid-refractory (SR) acute (a) and chronic (c) graft-versus-host disease (GVHD) cause high mortality. Preclinical evidence indicates the potent anti-inflammatory properties of the JAK1/2 inhibitor ruxolitinib. In this retrospective survey, 19 stem cell transplant centers in Europe and the United States reported outcome data from 95 patients who had received ruxolitinib as salvage therapy for SR-GVHD. Patients were classified as having SR-aGVHD (n=54, all grades III or IV) or SR-cGVHD (n=41, all moderate or severe). The median number of previous GVHD-therapies was 3 for both SR-aGVHD (1-7) and SR-cGVHD (1-10). The overall response rate was 81.5% (44/54) in SR-aGVHD including 25 complete responses (46.3%), while for SR-cGVHD the ORR was 85.4% (35/41). Of those patients responding to ruxolitinib, the rate of GVHD-relapse was 6.8% (3/44) and 5.7% (2/35) for SR-aGVHD and SR-cGVHD, respectively. The 6-month-survival was 79% (67.3-90.7%, 95% confidence interval (CI)) and 97.4% (92.3-100%, 95% CI) for SR-aGVHD and SR-cGVHD, respectively. Cytopenia and cytomegalovirus-reactivation were observed during ruxolitinib treatment in both SR-aGVHD (30/54, 55.6% and 18/54, 33.3%) and SR-cGVHD (7/41, 17.1% and 6/41, 14.6%) patients. Ruxolitinib may constitute a promising new treatment option for SR-aGVHD and SR-cGVHD that should be validated in a prospective trial.
Collapse
Affiliation(s)
- R Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - A Burchert
- Department of Hematology, Oncology and Immunology, Philipps University Marburg, and University Medical Center Giessen and Marburg, Marburg, Germany
| | - C Lengerke
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - M Verbeek
- III Department of Internal Medicine, Technical University of Munich, Munich, Germany
| | - K Maas-Bauer
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - S K Metzelder
- Department of Hematology, Oncology and Immunology, Philipps University Marburg, and University Medical Center Giessen and Marburg, Marburg, Germany
| | - S Spoerl
- III Department of Internal Medicine, Technical University of Munich, Munich, Germany
| | - M Ditschkowski
- Department of Bone Marrow Transplantation, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - M Ecsedi
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - K Sockel
- Department of Hematology and Oncology, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - F Ayuk
- Department of Stem Cell Transplantation, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - S Ajib
- Department of Internal Medicine II, University Hospital, Frankfurt/Main, Germany
| | - F S de Fontbrune
- Hematology Stem cell transplant Unit, Saint Louis Hospital, APHP, Paris, France
| | - I-K Na
- Department of Stem Cell Transplantation, Charité University Medicine Berlin, Berlin, Germany
| | - L Penter
- Department of Stem Cell Transplantation, Charité University Medicine Berlin, Berlin, Germany
| | - U Holtick
- Department of Internal Medicine I, University Hospital Cologne, Cologne, Germany
| | - D Wolf
- Medical Clinic III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - E Schuler
- Department of Hematology, Oncology and Clinical Immunology, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - E Meyer
- Department of Bone Marrow Transplantation, Stanford University Medical School, Stanford, CA, USA
| | - P Apostolova
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - H Bertz
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - R Marks
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - M Lübbert
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - R Wäsch
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - C Scheid
- Department of Internal Medicine I, University Hospital Cologne, Cologne, Germany
| | - F Stölzel
- Department of Hematology and Oncology, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - R Ordemann
- Department of Hematology and Oncology, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - G Bug
- Department of Internal Medicine II, University Hospital, Frankfurt/Main, Germany
| | - G Kobbe
- Department of Hematology, Oncology and Clinical Immunology, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - R Negrin
- Department of Bone Marrow Transplantation, Stanford University Medical School, Stanford, CA, USA
| | - M Brune
- Department of Internal Medicine, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - A Spyridonidis
- Department of Bone Marrow Transplantation, Patras University Medical School, Patras, Greece
| | - A Schmitt-Gräff
- Department of Pathology, Freiburg University Medical Center, Freiburg, Germany
| | | | - G Huls
- Radboud University Medical Centre, Nijmegen, The Netherlands
| | - S Mielke
- Department of Hematology and Oncology, University Medical Centre Würzburg, Würzburg, Germany
| | - G U Grigoleit
- Department of Hematology and Oncology, University Medical Centre Würzburg, Würzburg, Germany
| | - J Kuball
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R Flynn
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - G Ihorst
- Clinical Trials Unit, Department of Hematology, Freiburg University Medical Center, Freiburg, Germany
| | - J Du
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - B R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - R Arnold
- Department of Stem Cell Transplantation, Charité University Medicine Berlin, Berlin, Germany
| | - N Kröger
- Department of Stem Cell Transplantation, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - J Passweg
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - J Halter
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - G Socié
- Hematology Stem cell transplant Unit, Saint Louis Hospital, APHP, Paris, France
| | - D Beelen
- Department of Bone Marrow Transplantation, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - C Peschel
- III Department of Internal Medicine, Technical University of Munich, Munich, Germany
| | - A Neubauer
- Department of Hematology, Oncology and Immunology, Philipps University Marburg, and University Medical Center Giessen and Marburg, Marburg, Germany
| | - J Finke
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - J Duyster
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| | - N von Bubnoff
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg, Germany
| |
Collapse
|
22
|
Politikos I, Kim HT, Nikiforow S, Li L, Brown J, Antin JH, Cutler C, Ballen K, Ritz J, Boussiotis VA. IL-7 and SCF Levels Inversely Correlate with T Cell Reconstitution and Clinical Outcomes after Cord Blood Transplantation in Adults. PLoS One 2015; 10:e0132564. [PMID: 26177551 PMCID: PMC4503696 DOI: 10.1371/journal.pone.0132564] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/11/2015] [Indexed: 11/19/2022] Open
Abstract
Recovery of thymopoiesis is critical for immune reconstitution after HSCT. IL-7 and SCF are two major thymotropic cytokines. We investigated whether the kinetics of circulating levels of these cytokines might provide insight into the prolonged immunodeficiency after double umbilical cord blood transplantation (dUCBT) in adults. We examined plasma levels of IL-7 and SCF, T-cell receptor rearrangement excision circle (TREC) levels and T cell subsets in 60 adult patients undergoing dUCBT. Median levels of IL-7 increased by more than 3-fold at 4 weeks and remained elevated through 100 days after dUCBT. SCF showed a less than 2-fold increase and more protracted elevation than IL-7. IL-7 levels inversely correlated with the reconstitution of various T cell subsets but not with TRECs. SCF levels inversely correlated with reconstitution of CD4+T cells, especially the naïve CD4+CD45RA+ subset, and with TRECs suggesting that SCF but not IL-7 had an effect on thymic regeneration. In Cox models, elevated levels of IL-7 and SCF were associated with higher non-relapse mortality (p = 0.03 and p = 0.01) and worse overall survival (p = 0.002 and p = 0.001). Elevated IL-7 but not SCF was also associated with development of GvHD (p = 0.03). Thus, IL-7 and SCF are elevated for a prolonged period after dUCBT and persistently high levels of these cytokines may correlate with worse clinical outcomes.
Collapse
Affiliation(s)
- Ioannis Politikos
- Department of Medicine and Division of Hematology Oncology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Haesook T. Kim
- Department of Computational Biology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Sarah Nikiforow
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Lequn Li
- Department of Medicine and Division of Hematology Oncology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Julia Brown
- Department of Medicine and Division of Hematology Oncology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Joseph H. Antin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Corey Cutler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Karen Ballen
- Bone Marrow Transplantation Unit, Massachusetts General Hospital, Boston, MA, United States of America
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Vassiliki A. Boussiotis
- Department of Medicine and Division of Hematology Oncology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
23
|
Kielsen K, Jordan KK, Uhlving HH, Pontoppidan PL, Shamim Z, Ifversen M, Heilmann C, Nielsen CH, Sengeløv H, Ryder LP, Müller KG. T cell reconstitution in allogeneic haematopoietic stem cell transplantation: prognostic significance of plasma interleukin-7. Scand J Immunol 2015; 81:72-80. [PMID: 25263171 DOI: 10.1111/sji.12244] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/18/2014] [Indexed: 12/25/2022]
Abstract
Infections and acute graft-versus-host disease (aGVHD) are major causes of treatment-related mortality and morbidity following allogeneic haematopoietic stem cell transplantation (HSCT). Both complications depend on reconstitution of the T-lymphocyte population based on donor T cells. Although it is well established that Interleukin-7 (IL-7) is a cytokine essential for de novo T cell development in the thymus and homoeostatic peripheral expansion of T cells, associations between circulating levels of IL-7 and T cell reconstitution following HSCT have not been investigated previously. We prospectively measured IL-7 levels in 81 patients undergoing myeloablative HSCT with either sibling donor or an unrelated donor. Plasma IL-7 levels peaked at day +7 post-transplant (1.3-82.4 pg/ml), at the time of maximal lymphopaenia. In multivariate analysis, peak levels of IL-7 were significantly higher in patients treated with anti-thymocyte globulin (ATG) compared with those not treated with ATG (P = 0.0079). IL-7 levels at day +7 were negatively associated with T cell counts at day +30 to +60 (at day +60: CD3(+) : β = -10.6 × 10(6) cells/l, P = 0.0030; CD8(+) : β = -8.4 × 10(6) cells/l, P = 0.061; CD4(+) : β = -2.1 × 10(6) cells/l, P = 0.062) in multivariate analyses. In adults, high IL-7 levels were associated with increased risk of grade II-IV aGVHD (OR = 5.4, P = 0.036) and reduced overall survival (P = 0.046). The present data indicate that high plasma levels of IL-7 in the early post-transplant period are predictive for slow T cell reconstitution, increased risk of aGVHD and increased mortality following HSCT.
Collapse
Affiliation(s)
- K Kielsen
- Institute for Inflammation Research, Department of Infectious Diseases and Rheumatology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Peripheral Blood CD38 Bright CD8+ Effector Memory T Cells Predict Acute Graft-versus-Host Disease. Biol Blood Marrow Transplant 2015; 21:1215-22. [PMID: 25881755 DOI: 10.1016/j.bbmt.2015.04.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/07/2015] [Indexed: 02/08/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is mediated by allogeneic T cell responses. We hypothesized that increases of peripheral blood-activated CD8+ effector memory T (TEM) cells would be observed after hematopoietic stem cell transplantation (HSCT) before onset of aGVHD symptoms. Blood was collected twice weekly after HSCT for 7 weeks in 49 consecutive pediatric and adult HSCT recipients. Samples were incubated with fluorochrome-conjugated antibodies against CD45, CD3, CD8, CD38, CD45RA, and CCR7 and analyzed using flow cytometry. TEM cells were defined as CD3+ CD8+ CCR7- CD45RA(-) lymphocytes. CD38 expression was used as a marker of T cell activation. Patients were followed for 100 days for development of aGVHD. Twenty-three patients developed grade 1 to 4 aGVHD at a median of 37 days (range, 15 to 79 days) after HCST. Absolute CD38 bright CD8+ TEM of > 35 cells/μL predicted aGVHD at a median of 8 days (range, 1 to 34) before aGVHD onset with a sensitivity of 82.6% and specificity of 91.6%. The cumulative incidence of aGVHD was 90% in patients with absolute CD38 bright CD8+ TEM >35 cells/μL and 15% in patients without (P < .0001). Quantification of CD38 bright CD8+ TEM cells may predict aGVHD in children and young adult HSCT recipients.
Collapse
|
25
|
CMV reactivation drives posttransplant T-cell reconstitution and results in defects in the underlying TCRβ repertoire. Blood 2015; 125:3835-50. [PMID: 25852054 DOI: 10.1182/blood-2015-03-631853] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 03/26/2015] [Indexed: 12/21/2022] Open
Abstract
Although cytomegalovirus (CMV) reactivation has long been implicated in posttransplant immune dysfunction, the molecular mechanisms that drive this phenomenon remain undetermined. To address this, we combined multiparameter flow cytometric analysis and T-cell subpopulation sorting with high-throughput sequencing of the T-cell repertoire, to produce a thorough evaluation of the impact of CMV reactivation on T-cell reconstitution after unrelated-donor hematopoietic stem cell transplant. We observed that CMV reactivation drove a >50-fold specific expansion of Granzyme B(high)/CD28(low)/CD57(high)/CD8(+) effector memory T cells (Tem) and resulted in a linked contraction of all naive T cells, including CD31(+)/CD4(+) putative thymic emigrants. T-cell receptor β (TCRβ) deep sequencing revealed a striking contraction of CD8(+) Tem diversity due to CMV-specific clonal expansions in reactivating patients. In addition to querying the topography of the expanding CMV-specific T-cell clones, deep sequencing allowed us, for the first time, to exhaustively evaluate the underlying TCR repertoire. Our results reveal new evidence for significant defects in the underlying CD8 Tem TCR repertoire in patients who reactivate CMV, providing the first molecular evidence that, in addition to driving expansion of virus-specific cells, CMV reactivation has a detrimental impact on the integrity and heterogeneity of the rest of the T-cell repertoire. This trial was registered at www.clinicaltrials.gov as #NCT01012492.
Collapse
|
26
|
Hannon M, Beguin Y, Ehx G, Servais S, Seidel L, Graux C, Maertens J, Kerre T, Daulne C, de Bock M, Fillet M, Ory A, Willems E, Gothot A, Humblet-Baron S, Baron F. Immune Recovery after Allogeneic Hematopoietic Stem Cell Transplantation Following Flu-TBI versus TLI-ATG Conditioning. Clin Cancer Res 2015; 21:3131-9. [PMID: 25779951 DOI: 10.1158/1078-0432.ccr-14-3374] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/06/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE A conditioning regimen for allogeneic hematopoietic cell transplantation (HCT) combining total lymphoid irradiation (TLI) plus anti-thymocyte globulin (ATG) has been developed to induce graft-versus-tumor effects without graft-versus-host disease (GVHD). EXPERIMENTAL DESIGN We compared immune recovery in 53 patients included in a phase II randomized study comparing nonmyeloablative HCT following either fludarabine plus 2 Gy total body irradiation (TBI arm, n = 28) or 8 Gy TLI plus ATG (TLI arm, n = 25). RESULTS In comparison with TBI patients, TLI patients had a similarly low 6-month incidence of grade II-IV acute GVHD, a lower incidence of moderate/severe chronic GVHD (P = 0.02), a higher incidence of CMV reactivation (P < 0.001), and a higher incidence of relapse (P = 0.01). While recovery of total CD8(+) T cells was similar in the two groups, with median CD8(+) T-cell counts reaching the normal values 40 to 60 days after allo-HCT, TLI patients had lower percentages of naïve CD8 T cells. Median CD4(+) T-cell counts did not reach the lower limit of normal values the first year after allo-HCT in the two groups. Furthermore, CD4(+) T-cell counts were significantly lower in TLI than in TBI patients the first 6 months after transplantation. Interestingly, while median absolute regulatory T-cell (Treg) counts were comparable in TBI and TLI patients, Treg/naïve CD4(+) T-cell ratios were significantly higher in TLI than in TBI patients the 2 first years after transplantation. CONCLUSIONS Immune recovery differs substantially between these two conditioning regimens, possibly explaining the different clinical outcomes observed (NCT00603954).
Collapse
Affiliation(s)
- Muriel Hannon
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium.
| | - Yves Beguin
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium. Department of Clinical Hematology, CHU of Liège, Liège, Belgium
| | - Grégory Ehx
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Sophie Servais
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium. Department of Clinical Hematology, CHU of Liège, Liège, Belgium
| | - Laurence Seidel
- Department of statistics, SIME, CHU of Liège, Liège, Belgium
| | - Carlos Graux
- Mont-Godine University Hospital (UCL), Yvoir, Belgium
| | | | | | - Coline Daulne
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Muriel de Bock
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Marianne Fillet
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Aurélie Ory
- Department of Clinical Hematology, CHU of Liège, Liège, Belgium
| | - Evelyne Willems
- Department of Clinical Hematology, CHU of Liège, Liège, Belgium
| | - André Gothot
- Department of Laboratory Medicine, University of Liège, Liège, Belgium
| | - Stéphanie Humblet-Baron
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Frédéric Baron
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium. Department of Clinical Hematology, CHU of Liège, Liège, Belgium
| |
Collapse
|
27
|
|
28
|
Paczesny S, Duncan C, Jacobsohn D, Krance R, Leung K, Carpenter P, Bollard C, Renbarger J, Cooke K. Opportunities and challenges of proteomics in pediatric patients: circulating biomarkers after hematopoietic stem cell transplantation as a successful example. Proteomics Clin Appl 2014; 8:837-50. [PMID: 25196024 DOI: 10.1002/prca.201400033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/30/2014] [Accepted: 09/03/2014] [Indexed: 11/06/2022]
Abstract
Biomarkers have the potential to improve diagnosis and prognosis, facilitate-targeted treatment, and reduce health care costs. Thus, there is great hope that biomarkers will be integrated in all clinical decisions in the near future. A decade ago, the biomarker field was launched with great enthusiasm because MS revealed that blood contains a rich library of candidate biomarkers. However, biomarker research has not yet delivered on its promise due to several limitations: (i) improper sample handling and tracking as well as limited sample availability in the pediatric population, (ii) omission of appropriate controls in original study designs, (iii) lability and low abundance of interesting biomarkers in blood, and (iv) the inability to mechanistically tie biomarker presence to disease biology. These limitations as well as successful strategies to overcome them are discussed in this review. Several advances in biomarker discovery and validation have been made in hematopoietic stem cell transplantation, the current most effective tumor immunotherapy, and these could serve as examples for other conditions. This review provides fresh optimism that biomarkers clinically relevant in pediatrics are closer to being realized based on: (i) a uniform protocol for low-volume blood collection and preservation, (ii) inclusion of well-controlled independent cohorts, (iii) novel technologies and instrumentation with low analytical sensitivity, and (iv) integrated animal models for exploring potential biomarkers and targeted therapies.
Collapse
Affiliation(s)
- Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Therapeutic activity of multiple common γ-chain cytokine inhibition in acute and chronic GVHD. Blood 2014; 125:570-80. [PMID: 25352130 DOI: 10.1182/blood-2014-06-581793] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The common γ chain (CD132) is a subunit of the interleukin (IL) receptors for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Because levels of several of these cytokines were shown to be increased in the serum of patients developing acute and chronic graft-versus-host disease (GVHD), we reasoned that inhibition of CD132 could have a profound effect on GVHD. We observed that anti-CD132 monoclonal antibody (mAb) reduced acute GVHD potently with respect to survival, production of tumor necrosis factor, interferon-γ, and IL-6, and GVHD histopathology. Anti-CD132 mAb afforded protection from GVHD partly via inhibition of granzyme B production in CD8 T cells, whereas exposure of CD8 T cells to IL-2, IL-7, IL-15, and IL-21 increased granzyme B production. Also, T cells exposed to anti-CD132 mAb displayed a more naive phenotype in microarray-based analyses and showed reduced Janus kinase 3 (JAK3) phosphorylation upon activation. Consistent with a role of JAK3 in GVHD, Jak3(-/-) T cells caused less severe GVHD. Additionally, anti-CD132 mAb treatment of established chronic GVHD reversed liver and lung fibrosis, and pulmonary dysfunction characteristic of bronchiolitis obliterans. We conclude that acute GVHD and chronic GVHD, caused by T cells activated by common γ-chain cytokines, each represent therapeutic targets for anti-CD132 mAb immunomodulation.
Collapse
|
30
|
Graft-versus-host disease biomarkers: omics and personalized medicine. Int J Hematol 2014; 98:275-92. [PMID: 23959582 DOI: 10.1007/s12185-013-1406-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 07/29/2013] [Indexed: 02/03/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the most effective form of tumor immunotherapy available to date and the frequency of transplants continues to increase worldwide. However, while allo-HSCT usually induces a beneficial graft-versus leukemia effect, a major source of morbidity and mortality following allo-HSCT is graft-versus-host disease (GVHD). Currently available diagnostic and staging tools frequently fail to identify those at higher risk for GVHD morbidity, treatment unresponsiveness, and death. Furthermore, there are shortcomings in the risk stratification of patients before GVHD clinical signs develop. In parallel, recent years have been characterized by an explosive evolution of omics technologies, largely due to technological advancements in chemistry, engineering, and bioinformatics. Building on these opportunities, plasma biomarkers have been identified and validated as promising diagnostic and prognostic tools for acute GVHD. This review summarizes current information on the types of GVHD biomarkers, the omics tools used to identify them, the biomarkers currently validated as acute GVHD markers, and future recommendations for incorporating biomarkers into new grading algorithms for risk-stratifying patients and creating more personalized treatment courses. Future directions will include randomized evaluations of these biomarkers in multicenter prospective studies while extending on the need for biomarkers of chronic GVHD.
Collapse
|
31
|
De Bock M, Beguin Y, Leprince P, Willems E, Baron F, Deroyer C, Seidel L, Cavalier E, de Seny D, Malaise M, Gothot A, Merville MP, Fillet M. Comprehensive plasma profiling for the characterization of graft-versus-host disease biomarkers. Talanta 2014; 125:265-75. [DOI: 10.1016/j.talanta.2014.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 02/28/2014] [Accepted: 03/11/2014] [Indexed: 02/07/2023]
|
32
|
Poiret T, Rane L, Remberger M, Omazic B, Gustafsson-Jernberg A, Vudattu NK, Ahmed R, Ernberg I, Winiarski J, Magalhaes I, Ringden O, Maeurer M. Reduced plasma levels of soluble interleukin-7 receptor during graft-versus-host disease (GVHD) in children and adults. BMC Immunol 2014; 15:25. [PMID: 24946690 PMCID: PMC4074150 DOI: 10.1186/1471-2172-15-25] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 05/09/2014] [Indexed: 11/10/2022] Open
Abstract
Background Interleukin 7 (IL-7) signals via the IL-7 receptor (IL-7R) and drives homeostatic T-cell proliferation in patients after allogeneic hematopoietic stem cell transplantation (aHSCT). Purpose We performed a prospective study in adults (n = 33) and children (n = 29) undergoing aHSCT measuring plasma IL-7 and soluble IL-7R (sIL-7R) concentrations between 1 and 12 months after HSCT in order to investigate the link between sIL-7R and clinical events after aHSCT. Results sIL-7R, but not IL-7, increased with time after HSCT in plasma from all patients enrolled in the study. sIL-7R values were higher at 2, 3, and 6 months (p < 0.01) if the donor was a sibling as compared to an unrelated donor. Increased sIL-7R levels were also identified in plasma from patients who were not treated with anti-thymocyte globulin (ATG). Low sIL-7R was associated with any grade of acute graft-versus-host disease (GVHD) at 2 and 6 months (p = 0.02) and with a positive CMV PCR at 2 months after HSCT (p < 0.05). Patients with cytomegalovirus (CMV) reactivation had increased IL-7 values at 2 and 3 months (p = 0.02) after HSCT. In multivariate analysis, lower sIL-7R levels were associated with acute GVHD (relative hazard (RH): 0.70, p > 0.01) and sibling donors (RH: 2.23, p = 0.004). Recipients of sibling grafts showed high levels of IL-7 (RH: 1.38, p < 0.05) and bone marrow recipients had low IL-7 levels (RH: 0.73, p = 0.04). Conclusions Measurement of the sIL-7R/IL-7 axis will help in guided immune monitoring after HSCT and guided interference with sIL-7R may be explored in GVHD management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Markus Maeurer
- Division of Therapeutic Immunology, Labmed, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Glauzy S, André-Schmutz I, Larghero J, Ezine S, de Latour RP, Moins-Teisserenc H, Servais S, Robin M, Socié G, Clave E, Toubert A. CXCR4-related increase of circulating human lymphoid progenitors after allogeneic hematopoietic stem cell transplantation. PLoS One 2014; 9:e91492. [PMID: 24621606 PMCID: PMC3951398 DOI: 10.1371/journal.pone.0091492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/10/2014] [Indexed: 01/05/2023] Open
Abstract
Immune recovery after profound lymphopenia is a major challenge in many clinical situations, such as allogeneic hematopoietic stem cell transplantation (allo-HSCT). Recovery depends, in a first step, on hematopoietic lymphoid progenitors production in the bone marrow (BM). In this study, we characterized CD34+Lin-CD10+ lymphoid progenitors in the peripheral blood of allo-HSCT patients. Our data demonstrate a strong recovery of this population 3 months after transplantation. This rebound was abolished in patients who developed acute graft-versus-host disease (aGVHD). A similar recovery profile was found for both CD24+ and CD24- progenitor subpopulations. CD34+lin-CD10+CD24- lymphoid progenitors sorted from allo-HSCT patients preserved their T cell potentiel according to in vitro T-cell differentiation assay and the expression profile of 22 genes involved in T-cell differentiation and homing. CD34+lin-CD10+CD24- cells from patients without aGVHD had reduced CXCR4 gene expression, consistent with an enhanced egress from the BM. CCR7 gene expression was reduced in patients after allo-HSCT, as were its ligands CCL21 and CCL19. This reduction was particularly marked in patients with aGVHD, suggesting a possible impact on thymic homing. Thus, the data presented here identify this population as an important early step in T cell reconstitution in humans and so, an important target when seeking to enhance immune reconstitution.
Collapse
Affiliation(s)
- Salomé Glauzy
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Isabelle André-Schmutz
- INSERM U768, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Jérôme Larghero
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Unité de Thérapie Cellulaire et CIC de Biothérapies, Hôpital Saint-Louis, AP-HP, Paris, France
| | | | - Régis Peffault de Latour
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- Service d'Hématologie-Greffe de Moelle, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Hélène Moins-Teisserenc
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Sophie Servais
- Service d'Hématologie-Greffe de Moelle, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Marie Robin
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- Service d'Hématologie-Greffe de Moelle, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Gérard Socié
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Service d'Hématologie-Greffe de Moelle, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Emmanuel Clave
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Antoine Toubert
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| |
Collapse
|
34
|
Matsuoka KI, Koreth J, Kim HT, Bascug G, McDonough S, Kawano Y, Murase K, Cutler C, Ho VT, Alyea EP, Armand P, Blazar BR, Antin JH, Soiffer RJ, Ritz J. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med 2013; 5:179ra43. [PMID: 23552371 DOI: 10.1126/scitranslmed.3005265] [Citation(s) in RCA: 372] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CD4(+)Foxp3(+) regulatory T cells (Tregs) play a central role in the maintenance of immune tolerance after allogeneic hematopoietic stem cell transplantation. We recently reported that daily administration of low-dose interleukin-2 (IL-2) induces selective expansion of functional Tregs and clinical improvement of chronic graft-versus-host disease (GVHD). To define the mechanisms of action of IL-2 therapy, we examined the immunologic effects of this treatment on homeostasis of CD4(+) T cell subsets after transplant. We first demonstrated that chronic GVHD is characterized by constitutive phosphorylation of signal transducer and activator of transcription 5 (Stat5) in conventional CD4(+) T cells (Tcons) associated with elevated amounts of IL-7 and IL-15 and relative functional deficiency of IL-2. IL-2 therapy resulted in the selective increase of Stat5 phosphorylation in Tregs and a decrease of phosphorylated Stat5 in Tcons. Over an 8-week period, IL-2 therapy induced a series of changes in Treg homeostasis, including increased proliferation, increased thymic export, and enhanced resistance to apoptosis. Low-dose IL-2 had minimal effects on Tcons. These findings define the mechanisms whereby low-dose IL-2 therapy restores the homeostasis of CD4(+) T cell subsets and promotes the reestablishment of immune tolerance.
Collapse
Affiliation(s)
- Ken-ichi Matsuoka
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kinetics of IL-7 and IL-15 levels after allogeneic peripheral blood stem cell transplantation following nonmyeloablative conditioning. PLoS One 2013; 8:e55876. [PMID: 23437070 PMCID: PMC3578874 DOI: 10.1371/journal.pone.0055876] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 01/03/2013] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND We analysed kinetics of IL-7 and IL-15 levels in 70 patients given peripheral blood stem cells after nonmyeloablative conditioning. METHODS EDTA-anticoagulated plasma and serum samples were obtained before conditioning and about once per week after transplantation until day 100. Samples were aliquoted and stored at -80°C within 3 hours after collection until measurement of cytokines. IL-7 and IL-15 levels were measured by ELISAs. RESULTS Median IL-7 plasma levels remained below 6 pg/L throughout the first 100 days, although IL-7 plasma levels were significantly higher on days 7 (5.1 pg/mL, P=0.002), 14 (5.2 pg/mL, P<0.001), and 28 (5.1 pg/mL, P=0.03) (but not thereafter) than before transplantation (median value of 3.8 pg/mL). Median IL-15 serum levels were significantly higher on days 7 (12.5 pg/mL, P<0.001), 14 (10.5 pg/mL, P<0.001), and 28 (6.2 pg/mL, P<0.001) than before transplantation (median value of 2.4 pg/mL). Importantly, IL-7 and IL-15 levels on days 7 or 14 after transplantation did not predict grade II-IV acute GVHD. CONCLUSIONS These data suggest that IL-7 and IL-15 levels remain relatively low after nonmyeloablative transplantation, and that IL-7 and IL-15 levels early after nonmyeloablative transplantation do not predict for acute GVHD.
Collapse
|
36
|
Abstract
Acute graft-versus-host disease (GVHD) is a leading cause of non-relapse mortality following allogeneic haematopoietic cell transplantation. Attempts to improve treatment response in clinically-established GVHD have not improved overall survival, often due to the increased risk of infectious complications. Alternative approaches to decrease GVHD-related morbidity and mortality have focused on the ability to predict GVHD prior to clinical manifestation in an effort to provide an opportunity to abort GVHD development, and to gain new insights into GVHD pathophysiology. This review outlines the research efforts to date that have identified clinical and laboratory-based factors that are predictive of acute GVHD and describes future directions in developing algorithms that will improve the ability to predict the development of clinically relevant GVHD.
Collapse
Affiliation(s)
- Andrew C Harris
- Blood and Marrow Transplant Program, Department of Pediatrics, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
37
|
Pratt LM, Liu Y, Ugarte-Torres A, Hoegh-Petersen M, Podgorny PJ, Lyon AW, Williamson TS, Khan FM, Chaudhry MA, Daly A, Stewart DA, Russell JA, Grigg A, Ritchie D, Storek J. IL15 levels on day 7 after hematopoietic cell transplantation predict chronic GVHD. Bone Marrow Transplant 2012; 48:722-8. [PMID: 23165502 DOI: 10.1038/bmt.2012.210] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic GVHD (cGVHD) is an important complication of allogeneic hematopoietic cell transplantation (HCT). As preemptive therapy might be efficacious if administered early post transplant, we set out to determine whether cGVHD can be predicted from the serum level of a biomarker on day 7 or 28. In a discovery cohort of 153 HCT recipients conditioned with BU, fludarabine and rabbit antithymocyte globulin (ATG), we determined serum levels of B-cell-activating factor, vascular endothelial growth factor, soluble TNF-α receptor 1, soluble IL2 receptor α, IL5, IL6, IL7, IL15, γ-glutamyl transpeptidase, cholinesterase, total protein, urea and ATG. Patients with low levels of IL15 (<30.6 ng/L) on day 7 had 2.7-fold higher likelihood of developing significant cGVHD (needing systemic immunosuppressive therapy) than patients with higher IL15 levels (P<0.001). This was validated in a validation cohort of 105 similarly-treated patients; those with low IL15 levels had 3.7-fold higher likelihood of developing significant cGVHD (P=0.001). Low IL15 was not associated with relapse; it trended to be associated with acute GVHD and was associated with low infection rates. In conclusion, low IL15 levels on day 7 are predictive of cGVHD, and thus could be useful in guiding preemptive therapy.
Collapse
Affiliation(s)
- L M Pratt
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 2012; 121:573-84. [PMID: 23160470 DOI: 10.1182/blood-2012-05-431718] [Citation(s) in RCA: 471] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Long-living memory stem T cells (T(SCM)) with the ability to self-renew and the plasticity to differentiate into potent effectors could be valuable weapons in adoptive T-cell therapy against cancer. Nonetheless, procedures to specifically target this T-cell population remain elusive. Here, we show that it is possible to differentiate in vitro, expand, and gene modify in clinically compliant conditions CD8(+) T(SCM) lymphocytes starting from naive precursors. Requirements for the generation of this T-cell subset, described as CD62L(+)CCR7(+)CD45RA(+)CD45R0(+)IL-7Rα(+)CD95(+), are CD3/CD28 engagement and culture with IL-7 and IL-15. Accordingly, T(SCM) accumulates early after hematopoietic stem cell transplantation. The gene expression signature and functional phenotype define this population as a distinct memory T-lymphocyte subset, intermediate between naive and central memory cells. When transplanted in immunodeficient mice, gene-modified naive-derived T(SCM) prove superior to other memory lymphocytes for the ability to expand and differentiate into effectors able to mediate a potent xenogeneic GVHD. Furthermore, gene-modified T(SCM) are the only T-cell subset able to expand and mediate GVHD on serial transplantation, suggesting self-renewal capacity in a clinically relevant setting. These findings provide novel insights into the origin and requirements for T(SCM) generation and pave the way for their clinical rapid exploitation in adoptive cell therapy.
Collapse
|
39
|
Hu B, Bao G, Zhang Y, Lin D, Wu Y, Wu D, Liu H. Donor NK Cells and IL-15 promoted engraftment in nonmyeloablative allogeneic bone marrow transplantation. THE JOURNAL OF IMMUNOLOGY 2012; 189:1661-70. [PMID: 22798668 DOI: 10.4049/jimmunol.1103199] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Donor NK cells could promote engraftment by suppressing host alloreactive responses during allogeneic bone marrow transplantation (allo-BMT). The biological activity of NK cells could be significantly enhanced by IL-15. The current study attempted to evaluate the effect of donor NK cells and IL-15 administration on engraftment and immune reconstitution in a murine nonmyeloablative allo-BMT model. Mice infused with donor NK cells and treated with IL-15 during nonmyeloablative allo-BMT resulted in increased donor engraftment compared with either treatment alone. The number of donor-derived cell subsets also increased in the spleen of the recipient mice with combination treatment. The alloreactivity to donor type Ags was significantly reduced in the recipient mice with donor NK cell infusion and IL-15 treatment, which was manifested by decreased proliferation and IL-2 secretion of splenocytes from recipient mice in response to donor type Ags in MLR and decreased capacity of the splenocytes killing donor type tumor targets. We subsequently exposed recipient mice to reduced irradiation conditioning and showed that donor NK cell infusion and hydrodynamic injection-mediated IL-15 expression could synergistically promote donor engraftment and suppress alloreactivity during nonmyeloablative allo-BMT. Infusion of CFSE-labeled donor CD45.1(+) NK cells demonstrated that IL-15 could enhance the infused donor NK cell proliferation and function in vivo. IL-15 treatment also promoted donor bone marrow-derived NK cell development and function. Thus, donor NK cell infusion and IL-15 treatment could synergistically promote the engraftment and the development of donor-derived cell subsets and suppress the host alloresponse in a murine nonmyeloablative allo-BMT model.
Collapse
Affiliation(s)
- Bo Hu
- Laboratory of Cellular and Molecular Tumor Immunology, Department of Hematology, Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
Barao I, Alvarez M, Redelman D, Weiss JM, Ortaldo JR, Wiltrout RH, Murphy WJ. Hydrodynamic delivery of human IL-15 cDNA increases murine natural killer cell recovery after syngeneic bone marrow transplantation. Biol Blood Marrow Transplant 2011; 17:1754-64. [PMID: 21906575 DOI: 10.1016/j.bbmt.2011.08.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 08/31/2011] [Indexed: 01/26/2023]
Abstract
Immune deficiency immediately following bone marrow transplantation (BMT) increases susceptibility to opportunistic infections as well as tumor relapse. Natural Killer (NK) cells play important roles in the resistance to virally infected and transformed cells. Interleukin (IL)-15 has been shown to be essential for NK cell development and survival. We administered human (h) IL-15 cDNA (pIL-15) via hydrodynamic delivery to murine recipients undergoing congenic BMT to determine its effects on NK cell reconstitution. Hydrodynamic pIL-15 delivery resulted in high levels of hIL-15 protein in the serum that lasted for several days and then quickly declined. The appearance of hIL-15 was followed by a significant increase of mature donor-derived NK cells within the bone marrow, spleens, and livers of the treated recipients. No accumulation of immature NK cell progenitors was observed. The NK cells from IL-15-treated recipients displayed an activated phenotype and were lytically active toward tumor targets in vitro to a similar degree as did those cells from recipients treated with control plasmid. This suggests that the predominant effect of IL-15 was a quantitative increase in total NK cell numbers and not qualitative changes in NK cell functions. No toxicities or adverse effects were observed. Studies performed in transplanted mice bearing renal carcinoma tumors demonstrated that this mode of hIL-15 gene delivery resulted in increased antitumor responses. These results support the use of cytokine gene transfer-based regimens as a platform to augment NK cell recovery after BMT.
Collapse
Affiliation(s)
- Isabel Barao
- University of Nevada, Reno, Department of Microbiology and Immunology, Reno, Nevada, USA
| | | | | | | | | | | | | |
Collapse
|