1
|
Mir M, Faiz S, Bommakanti AG, Sheshadri A. Pulmonary Immunocompromise in Stem Cell Transplantation and Cellular Therapy. Clin Chest Med 2025; 46:129-147. [PMID: 39890284 DOI: 10.1016/j.ccm.2024.10.010] [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: 02/03/2025]
Abstract
Hematopoietic cell transplantation (HCT) and cellular therapies, such as chimeric-antigen receptor T-cell (CAR-T) treatments, are potentially curative treatments for certain hematologic malignancies and some nonmalignant disorders. However, pulmonary complications, both infectious and noninfectious, remain a significant cause of morbidity and mortality in patients who receive cellular therapies. This review article provides an overview of pulmonary complications encountered in the context of HCT and CAR-T. The authors discuss mechanisms of underlying immunocompromise that lead to a rise in infections. Additionally, they highlight key noninfectious complications of HCT that can mimic acute infections and suggest diagnostic approaches and preventive strategies to distinguish these entities promptly.
Collapse
Affiliation(s)
- Mahnoor Mir
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School at UTHealth, Houston, TX 77030, USA; Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Saadia Faiz
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anuradha G Bommakanti
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School at UTHealth, Houston, TX 77030, USA; Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Roux HM, Marouf A, Dutrieux J, Charmeteau-De Muylder B, Figueiredo-Morgado S, Avettand-Fenoel V, Cuvelier P, Naudin C, Bouaziz F, Geri G, Couëdel-Courteille A, Squara P, Marullo S, Cheynier R. Genetically determined thymic function affects strength and duration of immune response in COVID patients with pneumonia. SCIENCE ADVANCES 2023; 9:eadh7969. [PMID: 37738336 PMCID: PMC10516486 DOI: 10.1126/sciadv.adh7969] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/23/2023] [Indexed: 09/24/2023]
Abstract
Thymic activation improves the outcome of COVID-19 patients with severe pneumonia. The rs2204985 genetic polymorphism within the TCRA-TCRD locus, which affects thymic output in healthy individuals, was found here to modify SARS-CoV-2-specific immunity and disease severity in COVID-19 patients with severe pneumonia. Forty patients with severe COVID-19 pneumonia were investigated. The GG genotype at the rs2204985 locus was associated, independently of age and sex, with stronger and long-lasting anti-SARS-CoV-2 helper and cytotoxic T cell responses 6 months after recovery. The GG genotype was also associated with less severe lung involvement, higher thymic production, and higher counts of blood naïve T lymphocytes, including recent thymic emigrants, and a larger population of activated stem cell memory CD4+ T cells. Overall, GG patients developed a more robust and sustained immunity to SARS-CoV-2. Polymorphism at rs2204985 locus should be considered as an additional predictive marker of anti-SARS-CoV-2 immune response.
Collapse
Affiliation(s)
- Hélène M. Roux
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | - Amira Marouf
- Groupe Hospitalier privé Ambroise Paré-Hartmann, Département Recherche Innovation, 92200, Neuilly-Sur-Seine, France
| | - Jacques Dutrieux
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | | | | | - Véronique Avettand-Fenoel
- Université Paris Cité, Faculté de médecine, Institut Cochin-CNRS 8104/INSERM U1016 AP-HP, Service de Virologie, Hôpital Cochin, Paris, France
| | - Pelagia Cuvelier
- Groupe Hospitalier privé Ambroise Paré-Hartmann, Département Recherche Innovation, 92200, Neuilly-Sur-Seine, France
| | - Cécile Naudin
- Groupe Hospitalier privé Ambroise Paré-Hartmann, Département Recherche Innovation, 92200, Neuilly-Sur-Seine, France
| | - Fatma Bouaziz
- Groupe Hospitalier privé Ambroise Paré-Hartmann, Département Recherche Innovation, 92200, Neuilly-Sur-Seine, France
| | - Guillaume Geri
- Groupe Hospitalier privé Ambroise Paré-Hartmann, Département Recherche Innovation, 92200, Neuilly-Sur-Seine, France
| | | | - Pierre Squara
- Groupe Hospitalier privé Ambroise Paré-Hartmann, Département Recherche Innovation, 92200, Neuilly-Sur-Seine, France
| | - Stefano Marullo
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | - Rémi Cheynier
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| |
Collapse
|
3
|
Borrill R, Poulton K, Wynn R. Immunology of cord blood T-cells favors augmented disease response during clinical pediatric stem cell transplantation for acute leukemia. Front Pediatr 2023; 11:1232281. [PMID: 37780051 PMCID: PMC10534014 DOI: 10.3389/fped.2023.1232281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/22/2023] [Indexed: 10/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) has been an important and efficacious treatment for acute leukemia in children for over 60 years. It works primarily through the graft-vs.-leukemia (GVL) effect, in which donor T-cells and other immune cells act to eliminate residual leukemia. Cord blood is an alternative source of stem cells for transplantation, with distinct biological and immunological characteristics. Retrospective clinical studies report superior relapse rates with cord blood transplantation (CBT), when compared to other stem cell sources, particularly for patients with high-risk leukemia. Xenograft models also support the superiority of cord blood T-cells in eradicating malignancy, when compared to those derived from peripheral blood. Conversely, CBT has historically been associated with an increased risk of transplant-related mortality (TRM) and morbidity, particularly from infection. Here we discuss clinical aspects of CBT, the unique immunology of cord blood T-cells, their role in the GVL effect and future methods to maximize their utility in cellular therapies for leukemia, honing and harnessing their antitumor properties whilst managing the risks of TRM.
Collapse
Affiliation(s)
- Roisin Borrill
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kay Poulton
- Transplantation Laboratory, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Robert Wynn
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
4
|
Jarosch S, Köhlen J, Ghimire S, Orberg ET, Hammel M, Gaag D, Evert M, Janssen KP, Hiergeist A, Gessner A, Weber D, Meedt E, Poeck H, D'Ippolito E, Holler E, Busch DH. Multimodal immune cell phenotyping in GI biopsies reveals microbiome-related T cell modulations in human GvHD. Cell Rep Med 2023; 4:101125. [PMID: 37467715 PMCID: PMC10394271 DOI: 10.1016/j.xcrm.2023.101125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/13/2023] [Accepted: 06/22/2023] [Indexed: 07/21/2023]
Abstract
Acute graft-versus-host disease (aGvHD) is a significant complication after allogeneic hematopoietic stem cell transplantation (aHSCT), but major factors determining disease severity are not well defined yet. By combining multiplexed tissue imaging and single-cell RNA sequencing on gastrointestinal biopsies from aHSCT-treated individuals with fecal microbiome analysis, we link high microbiome diversity and the abundance of short-chain fatty acid-producing bacteria to the sustenance of suppressive regulatory T cells (Tregs). Furthermore, aGvHD severity strongly associates with the clonal expansion of mainly CD8 T cells, which we find distributed over anatomically distant regions of the gut, persistent over time, and inversely correlated with the presence of suppressive Tregs. Overall, our study highlights the pathophysiological importance of expanded CD8 T cell clones in the progression of aGvHD toward more severe clinical manifestations and strongly supports the further development of microbiome interventions as GvHD treatment via repopulation of the gut Treg niche to suppress inflammation.
Collapse
Affiliation(s)
- Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, 88397 Biberach an der Riß, Germany
| | - Jan Köhlen
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Sakhila Ghimire
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany
| | - Erik Thiele Orberg
- Department of Medicine III, Technical University of Munich (TUM), School of Medicine, Klinikum rechts der Isar TUM, 81675 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Monika Hammel
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Doris Gaag
- Institute for Pathology, University of Regensburg, 93053 Regensburg, Germany
| | - Matthias Evert
- Institute for Pathology, University of Regensburg, 93053 Regensburg, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Andreas Hiergeist
- Institute of Clinical Microbiology and Hygiene, University Medical Center, 93053 Regensburg, Germany
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Medical Center, 93053 Regensburg, Germany
| | - Daniela Weber
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany
| | - Elisabeth Meedt
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany
| | - Hendrik Poeck
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany; Leibniz Institute for Immuntherapie (LIT), Regensburg, Germany
| | - Elvira D'Ippolito
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Ernst Holler
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany.
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany.
| |
Collapse
|
5
|
Giglio F, Xue E, Barone A, Lorentino F, Greco R, Ruggeri A, Zambelli M, Parisi C, Milani R, Clerici D, Piemontese S, Marktel S, Lazzari L, Marcatti M, Bernardi M, Corti C, Lupo-Stanghellini MT, Ciceri F, Peccatori J. Intrabone transplant of a single unwashed umbilical cord blood unit with ATG-free and sirolimus-based GvHD prophylaxis: fast immune-reconstitution and long-term disease control in 30 patients with high-risk diseases. Transplant Cell Ther 2023:S2666-6367(23)01297-6. [PMID: 37244644 DOI: 10.1016/j.jtct.2023.05.015] [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: 03/03/2023] [Revised: 04/30/2023] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
INTRO Several strategies have been explored with the attempt of improving safety and feasibility of umbilical cord blood transplant (UCBT) in adults. AIM The aim of this retrospective analysis was to examine the safety and efficacy of intrabone transplant of a single unwashed cord blood unit in an ATG-free, sirolimus-based graft-versus-host prophylaxis platform. METHODS We collected data of all consecutive UCBT infused intrabone and unwashed at San Raffaele Hospital in Milan between 2012 and 2021. RESULTS Thirty-one consecutive UCBT were identified. All but 3 units had a high-resolution HLA typing on 8 loci at time of selection. At cryopreservation, the median number of CD34+ cells and total nucleated cells (TNCs) were 1 × 105/kg (0.6-12.0) and 2.8 × 107/kg (1.48-5.6), respectively. Eighty seven percent of patients received myeloablative conditioning; seventy seven percent of patients were transplanted for acute myeloid leukemia. Median follow-up among survivors was 38.2 months (range 10.4-123.6). No adverse events were related to the intrabone infusion at bedside under short-conscious peri-procedural sedation and to the no wash technique. After thawing, CD34+ and TNCs were 0.8 × 105/kg (0.1-2.3) and 1.42 × 107/kg (0.69-3.2) respectively. Median time to engraftment was 27 and 53 days for neutrophils and platelets, respectively; one patient rejected the transplant and was subsequently rescued with a salvage transplant. Median time to CD3+ above 100/μL was 30 days. 100-day CI of III-IV aGvHD was 12.9% (95%CI 4-27.3%), 2-year CI of moderate-to-severe chronic GvHD was 11.8% (95% CI 2.7-28.3%); at 2-year, OS was 52.7% (95%CI 33-69%), relapse incidence was 30.7% (95% CI 13.7-49.6%) and TRM of 29% (95%CI 14.3-45.6%). In univariate analysis CD34+ infused counts did not impact on transplant outcomes. In patients transplanted in first complete remission, relapse rate was 13% with an OS above 90% at 2 years. CONCLUSIONS Intrabone infusion of single CB unit was feasible, with no adverse reactions related to the no wash/intrabone infusion. We documented a low incidence of chronic GVHD and disease relapse with a fast immune-reconstitution.
Collapse
Affiliation(s)
- Fabio Giglio
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Elisabetta Xue
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Francesca Lorentino
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy; PhD Program in Public Health, Department of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Raffaella Greco
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Annalisa Ruggeri
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Matilde Zambelli
- Immunohematology and Transfusion Medicine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Cristina Parisi
- Immunohematology and Transfusion Medicine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Raffaella Milani
- Immunohematology and Transfusion Medicine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Daniela Clerici
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Simona Piemontese
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Sarah Marktel
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Lorenzo Lazzari
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Magda Marcatti
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Massimo Bernardi
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Consuelo Corti
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Fabio Ciceri
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Jacopo Peccatori
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
6
|
Watson SA, Javanmardi Y, Zanieri L, Shahreza S, Ragazzini R, Bonfanti P, Moeendarbary E. Integrated role of human thymic stromal cells in hematopoietic stem cell extravasation. Bioeng Transl Med 2023; 8:e10454. [PMID: 36925684 PMCID: PMC10013751 DOI: 10.1002/btm2.10454] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
The human thymus is the site of T-cell maturation and induction of central tolerance. Hematopoietic stem cell (HSC)-derived progenitors are recruited to the thymus from the fetal liver during early prenatal development and from bone marrow at later stages and postnatal life. The mechanism by which HSCs are recruited to the thymus is poorly understood in humans, though mouse models have indicated the critical role of thymic stromal cells (TSC). Here, we developed a 3D microfluidic assay based on human cells to model HSC extravasation across the endothelium into the extracellular matrix. We found that the presence of human TSC consisting of cultured thymic epithelial cells (TEC) and interstitial cells (TIC) increases the HSC extravasation rates by 3-fold. Strikingly, incorporating TEC or TIC alone is insufficient to perturb HSC extravasation rates. Furthermore, we identified complex gene expressions from interactions between endothelial cells, TEC and TIC modulates the HSCs extravasation. Our results suggest that comprehensive signaling from the complex thymic microenvironment is crucial for thymus seeding and that our system will allow manipulation of these signals with the potential to increase thymocyte migration in a therapeutic setting.
Collapse
Affiliation(s)
- Sara A. Watson
- Department of Mechanical EngineeringUCLLondonUK
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
| | | | - Luca Zanieri
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | | | - Roberta Ragazzini
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | - Paola Bonfanti
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | | |
Collapse
|
7
|
Dekker L, Sanders E, Lindemans CA, de Koning C, Nierkens S. Naive T Cells in Graft Versus Host Disease and Graft Versus Leukemia: Innocent or Guilty? Front Immunol 2022; 13:893545. [PMID: 35795679 PMCID: PMC9250980 DOI: 10.3389/fimmu.2022.893545] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
The outcome of allogeneic hematopoietic cell transplantation (allo-HCT) largely depends on the development and management of graft-versus-host disease (GvHD), infections, and the occurrence of relapse of malignancies. Recent studies showed a lower incidence of chronic GvHD and severe acute GvHD in patients receiving naive T cell depleted grafts compared to patients receiving complete T cell depleted grafts. On the other hand, the incidence of acute GvHD in patients receiving cord blood grafts containing only naive T cells is rather low, while potent graft-versus-leukemia (GvL) responses have been observed. These data suggest the significance of naive T cells as both drivers and regulators of allogeneic reactions. The naive T cell pool was previously thought to be a quiescent, homogenous pool of antigen-inexperienced cells. However, recent studies showed important differences in phenotype, differentiation status, location, and function within the naive T cell population. Therefore, the adequate recovery of these seemingly innocent T cells might be relevant in the imminent allogeneic reactions after allo-HCT. Here, an extensive review on naive T cells and their contribution to the development of GvHD and GvL responses after allo-HCT is provided. In addition, strategies specifically directed to stimulate adequate reconstitution of naive T cells while reducing the risk of GvHD are discussed. A better understanding of the relation between naive T cells and alloreactivity after allo-HCT could provide opportunities to improve GvHD prevention, while maintaining GvL effects to lower relapse risk.
Collapse
Affiliation(s)
- Linde Dekker
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Evy Sanders
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Coco de Koning
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
8
|
Rosichini M, Catanoso M, Screpanti I, Felli MP, Locatelli F, Velardi E. Signaling Crosstalks Drive Generation and Regeneration of the Thymus. Front Immunol 2022; 13:920306. [PMID: 35734178 PMCID: PMC9207182 DOI: 10.3389/fimmu.2022.920306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/17/2022] [Indexed: 12/19/2022] Open
Abstract
Optimal recovery of immune competence after periods of hematopoietic insults or stress is crucial to re-establish patient response to vaccines, pathogens and tumor antigens. This is particularly relevant for patients receiving high doses of chemotherapy or radiotherapy, who experience prolonged periods of lymphopenia, which can be associated with an increased risk of infections, malignant relapse, and adverse clinical outcome. While the thymus represents the primary organ responsible for the generation of a diverse pool of T cells, its function is profoundly impaired by a range of acute insults (including those caused by cytoreductive chemo/radiation therapy, infections and graft-versus-host disease) and by the chronic physiological deterioration associated with aging. Impaired thymic function increases the risk of infections and tumor antigen escape due to a restriction in T-cell receptor diversity and suboptimal immune response. Therapeutic approaches that can promote the renewal of the thymus have the potential to restore immune competence in patients. Previous work has documented the importance of the crosstalk between thymocytes and thymic epithelial cells in establishing correct architecture and function of thymic epithelium. This crosstalk is relevant not only during thymus organogenesis, but also to promote the recovery of its function after injuries. In this review, we will analyze the signals involved in the crosstalk between TECs and hematopoietic cells. We will focus in particular on how signals from T-cells can regulate TEC function and discuss the relevance of these pathways in restoring thymic function and T-cell immunity in experimental models, as well as in the clinical setting.
Collapse
Affiliation(s)
- Marco Rosichini
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marialuigia Catanoso
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Pia Felli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Maternal and Child Health, Sapienza University of Rome, Rome, Italy
| | - Enrico Velardi
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Enrico Velardi,
| |
Collapse
|
9
|
Somasundaram A, Cillo AR, Lampenfeld C, Workman CJ, Kunning S, Oliveri LN, Velez M, Joyce S, Calderon M, Dadey R, Rajasundaram D, Normolle DP, Watkins SC, Herman JG, Kirkwood JM, Lipson EJ, Ferris RL, Bruno TC, Vignali DAA. Systemic immune dysfunction in cancer patients driven by IL6 induction of LAG3 in peripheral CD8+ T cells. Cancer Immunol Res 2022; 10:885-899. [PMID: 35587532 DOI: 10.1158/2326-6066.cir-20-0736] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 06/10/2021] [Accepted: 05/17/2022] [Indexed: 11/16/2022]
Abstract
Many cancer patients do not develop a durable response to the current standard of care immunotherapies, despite substantial advances in targeting immune inhibitory receptors. A potential compounding issue, which may serve as an unappreciated, dominant resistance mechanism, is an inherent systemic immune dysfunction that is often associated with advanced cancer. Minimal response to inhibitory receptor (IR) blockade therapy and increased disease burden have been associated with peripheral CD8+ T-cell dysfunction, characterized by suboptimal T-cell proliferation and chronic expression of IRs (eg. Programmed Death 1 [PD1] and Lymphocyte Activation Gene 3 [LAG3]). Here, we demonstrated that approximately a third of cancer patients analyzed in this study have peripheral CD8+ T cells that expressed robust intracellular LAG3 (LAG3IC), but not surface LAG3 (LAG3SUR) due to A Disintegrin and Metalloproteinase domain-containing protein 10 (ADAM10) cleavage. This associated with poor disease prognosis and decreased CD8+ T-cell function, which could be partially reversed by anti-LAG3. Systemic immune dysfunction was restricted to CD8+ T cells, including, in some cases, a high percentage of peripheral naïve CD8+ T cells, and was driven by the cytokine IL6 via STAT3. These data suggest that additional studies are warrented to determine if the combination of increased LAG3IC in peripheral CD8+ T cells and elevated systemic IL6 can serve as predictive biomarkers and identify which cancer patients may benefit from LAG3 blockade.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maria Velez
- University of Pittsburgh, Pittsburgh, PA, United States
| | - Sonali Joyce
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Michael Calderon
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Rebekah Dadey
- University of Pittsburgh, Pittsburgh, PA, United States
| | | | | | | | | | | | - Evan J Lipson
- Johns Hopkins University School of Medicine, BALTIMORE, MD, United States
| | - Robert L Ferris
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States
| | - Tullia C Bruno
- University of Colorado Boulder, Pittsburgh, PA, United States
| | | |
Collapse
|
10
|
Itamura H, Shindo T, Muranushi H, Kitaura K, Okada S, Shin-I T, Suzuki R, Takaori-Kondo A, Kimura S. Pharmacological MEK inhibition promotes polyclonal T-cell reconstitution and suppresses xenogeneic GVHD. Cell Immunol 2021; 367:104410. [PMID: 34274730 DOI: 10.1016/j.cellimm.2021.104410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 11/21/2022]
Abstract
Rapid immune reconstitution without developing graft-versus-host disease (GVHD) is required for the success of allogeneic hematopoietic stem cell transplantation. Here, we analyzed the effects of pharmacological MEK inhibition on human polyclonal T-cell reconstitution in a humanized mouse GVHD model utilizing deep sequencing-based T-cell receptor (TCR) repertoire analysis. GVHD mice exhibited a skewed TCR repertoire with a common clone within target organs. The MEK inhibitor trametinib ameliorated GVHD and enabled engraftment of diverse T-cell clones. Furthermore, trametinib also ameliorated GVHD sparing diverse T cell repertoire, even when it was given from day 15 through 28. Although tacrolimus also reduced development of GVHD, it disturbed diverse T cell reconstitution and resulted in skewed TCR repertoire. Thus, trametinib not only suppresses GVHD-inducing T cells but also promotes human T cell reconstitution in vivo, providing a novel rationale for translational studies targeting human GVHD.
Collapse
Affiliation(s)
- Hidekazu Itamura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Takero Shindo
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan; Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Hiroyuki Muranushi
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | | | - Ryuji Suzuki
- Repertoire Genesis Inc., Ibaraki, Japan; Department of Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, Sagamihara, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
11
|
Velardi E, Clave E, Arruda LCM, Benini F, Locatelli F, Toubert A. The role of the thymus in allogeneic bone marrow transplantation and the recovery of the peripheral T-cell compartment. Semin Immunopathol 2021; 43:101-117. [PMID: 33416938 DOI: 10.1007/s00281-020-00828-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/14/2020] [Indexed: 12/11/2022]
Abstract
As the thymus represents the primary site of T-cell development, optimal thymic function is of paramount importance for the successful reconstitution of the adaptive immunity after allogeneic hematopoietic stem cell transplantation. Thymus involutes as part of the aging process and several factors, including previous chemotherapy treatments, conditioning regimen used in preparation to the allograft, occurrence of graft-versus-host disease, and steroid therapy that impair the integrity of the thymus, thus affecting its role in supporting T-cell neogenesis. Although the pathways governing its regeneration are still poorly understood, the thymus has a remarkable capacity to recover its function after damage. Measurement of both recent thymic emigrants and T-cell receptor excision circles is valuable tools to assess thymic output and gain insights on its function. In this review, we will extensively discuss available data on factors regulating thymic function after allogeneic hematopoietic stem cell transplantation, as well as the strategies and therapeutic approaches under investigation to promote thymic reconstitution and accelerate immune recovery in transplanted patients, including the use of cytokines, sex-steroid ablation, precursor T-cells, and thymus bioengineering. Although none of them is routinely used in the clinic, these approaches have the potential to enhance thymic function and immune recovery, not only in patients given an allograft but also in other conditions characterized by immune deficiencies related to a defective function of the thymus.
Collapse
Affiliation(s)
- Enrico Velardi
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy.
| | - Emmanuel Clave
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, F-75010, Paris, France
| | - Lucas C M Arruda
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Francesca Benini
- Department of Maternal and Child Health, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy.,Department of Maternal and Child Health, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Antoine Toubert
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, F-75010, Paris, France.,Laboratoire d'Immunologie et d'Histocompatibilité, AP-HP, Hopital Saint-Louis, F-75010, Paris, France
| |
Collapse
|
12
|
Yanir A, Schulz A, Lawitschka A, Nierkens S, Eyrich M. Immune Reconstitution After Allogeneic Haematopoietic Cell Transplantation: From Observational Studies to Targeted Interventions. Front Pediatr 2021; 9:786017. [PMID: 35087775 PMCID: PMC8789272 DOI: 10.3389/fped.2021.786017] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
Immune reconstitution (IR) after allogeneic haematopoietic cell transplantation (HCT) represents a central determinant of the clinical post-transplant course, since the majority of transplant-related outcome parameters such as graft-vs.-host disease (GvHD), infectious complications, and relapse are related to the velocity, quantity and quality of immune cell recovery. Younger age at transplant has been identified as the most important positive prognostic factor for favourable IR post-transplant and, indeed, accelerated immune cell recovery in children is most likely the pivotal contributing factor to lower incidences of GvHD and infectious complications in paediatric allogeneic HCT. Although our knowledge about the mechanisms of IR has significantly increased over the recent years, strategies to influence IR are just evolving. In this review, we will discuss different patterns of IR during various time points post-transplant and their impact on outcome. Besides IR patterns and cellular phenotypes, recovery of antigen-specific immune cells, for example virus-specific T cells, has recently gained increasing interest, as certain threshold levels of antigen-specific T cells seem to confer protection against severe viral disease courses. In contrast, the association between IR and a possible graft-vs. leukaemia effect is less well-understood. Finally, we will present current concepts of how to improve IR and how this could change transplant procedures in the near future.
Collapse
Affiliation(s)
- Asaf Yanir
- Bone Marrow Transplant Unit, Division of Haematology and Oncology, Schneider Children's Medical Center of Israel, Petach-Tikva, Israel.,The Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Anita Lawitschka
- St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria.,St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Matthias Eyrich
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University Medical Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
13
|
Ibrahim U, Keyzner A. Autoimmune hematologic complications of umbilical cord blood transplantation. Hematol Oncol Stem Cell Ther 2020; 14:104-109. [PMID: 32882204 DOI: 10.1016/j.hemonc.2020.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/13/2020] [Accepted: 07/26/2020] [Indexed: 10/23/2022] Open
Abstract
While umbilical cord blood is increasingly utilized as a stem cell source, immune complications associated with the procedure have been recognized. These complications result from significant immune system dysregulation and defective reconstitution following transplant causing an imbalance between T-cell subsets, aberrant B cells, and abnormal antibody production. This may occur up to 12 months after transplant coinciding with thymic regeneration in adults. The aim of our review is to describe the incidence, pathophysiology, clinical features, and prognosis of autoimmune cytopenias following umbilical cord blood transplant. Furthermore, we review the treatment strategies reported in the existing literature, describe the authors' experience with the complication, and highlight novel treatment options being studied. The knowledge of the occurrence and timing of autoimmune complications of umbilical cord blood transplantation is essential for detection and treatment of the disease. Emerging therapeutic options include interleukin-2 (IL-2), which is also being studied for the treatment of acute and chronic graft-versus-host disease. IL-2 has favorable effects on growth, differentiation, and function of regulatory T cells. Monoclonal antibody treatments, such as daratumumab, are also on the forefront and more experience with them will guide further treatment strategies.
Collapse
Affiliation(s)
- Uroosa Ibrahim
- Bone Marrow Transplant and Cellular Therapy Program, Tisch Cancer Institute, Mount Sinai Hospital, New York, USA.
| | - Alla Keyzner
- Bone Marrow Transplant and Cellular Therapy Program, Tisch Cancer Institute, Mount Sinai Hospital, New York, USA
| |
Collapse
|
14
|
Singh J, Mohtashami M, Anderson G, Zúñiga-Pflücker JC. Thymic Engraftment by in vitro-Derived Progenitor T Cells in Young and Aged Mice. Front Immunol 2020; 11:1850. [PMID: 32973763 PMCID: PMC7462002 DOI: 10.3389/fimmu.2020.01850] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
T cells play a critical role in mediating antigen-specific and long-term immunity against viral and bacterial pathogens, and their development relies on the highly specialized thymic microenvironment. T cell immunodeficiency can be acquired in the form of inborn errors, or can result from perturbations to the thymus due to aging or irradiation/chemotherapy required for cancer treatment. Hematopoietic stem cell transplant (HSCT) from compatible donors is a cornerstone for the treatment of hematological malignancies and immunodeficiency. Although it can restore a functional immune system, profound impairments exist in recovery of the T cell compartment. T cells remain absent or low in number for many months after HSCT, depending on a variety of factors including the age of the recipient. While younger patients have a shorter refractory period, the prolonged T cell recovery observed in older patients can lead to a higher risk of opportunistic infections and increased predisposition to relapse. Thus, strategies for enhancing T cell recovery in aged individuals are needed to counter thymic damage induced by radiation and chemotherapy toxicities, in addition to naturally occurring age-related thymic involution. Preclinical results have shown that robust and rapid long-term thymic reconstitution can be achieved when progenitor T cells, generated in vitro from HSCs, are co-administered during HSCT. Progenitor T cells appear to rely on lymphostromal crosstalk via receptor activator of NF-κB (RANK) and RANK-ligand (RANKL) interactions, creating chemokine-rich niches within the cortex and medulla that likely favor the recruitment of bone marrow-derived thymus seeding progenitors. Here, we employed preclinical mouse models to demonstrate that in vitro-generated progenitor T cells can effectively engraft involuted aged thymuses, which could potentially improve T cell recovery. The utility of progenitor T cells for aged recipients positions them as a promising cellular therapy for immune recovery and intrathymic repair following irradiation and chemotherapy, even in a post-involution thymus.
Collapse
Affiliation(s)
| | | | - Graham Anderson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
15
|
Dekker L, de Koning C, Lindemans C, Nierkens S. Reconstitution of T Cell Subsets Following Allogeneic Hematopoietic Cell Transplantation. Cancers (Basel) 2020; 12:E1974. [PMID: 32698396 PMCID: PMC7409323 DOI: 10.3390/cancers12071974] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023] Open
Abstract
Allogeneic (allo) hematopoietic cell transplantation (HCT) is the only curative treatment option for patients suffering from chemotherapy-refractory or relapsed hematological malignancies. The occurrence of morbidity and mortality after allo-HCT is still high. This is partly correlated with the immunological recovery of the T cell subsets, of which the dynamics and relations to complications are still poorly understood. Detailed information on T cell subset recovery is crucial to provide tools for better prediction and modulation of adverse events. Here, we review the current knowledge regarding CD4+ and CD8+ T cells, γδ T cells, iNKT cells, Treg cells, MAIT cells and naive and memory T cell reconstitution, as well as their relations to outcome, considering different cell sources and immunosuppressive therapies. We conclude that the T cell subsets reconstitute in different ways and are associated with distinct adverse and beneficial events; however, adequate reconstitution of all the subsets is associated with better overall survival. Although the exact mechanisms involved in the reconstitution of each T cell subset and their associations with allo-HCT outcome need to be further elucidated, the data and suggestions presented here point towards the development of individualized approaches to improve their reconstitution. This includes the modulation of immunotherapeutic interventions based on more detailed immune monitoring, aiming to improve overall survival changes.
Collapse
Affiliation(s)
- Linde Dekker
- Princess Máxima Center for Pediatric Oncology, Utrecht University, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (L.D.); (C.L.)
| | - Coco de Koning
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Caroline Lindemans
- Princess Máxima Center for Pediatric Oncology, Utrecht University, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (L.D.); (C.L.)
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht University, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (L.D.); (C.L.)
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| |
Collapse
|
16
|
Han FF, Fan H, Ren LL, Wang HG, Wang C, Ma X, Liu LH, He Q, Guo CL. Profiling the pattern of human TRB/IGH-CDR3 repertoire in liver transplantation patients via high-throughput sequencing analysis. Scand J Immunol 2020; 92:e12912. [PMID: 32458431 DOI: 10.1111/sji.12912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/29/2020] [Accepted: 05/19/2020] [Indexed: 12/15/2022]
Abstract
Immune processes in liver transplantation remain poorly understood. Acute allograft rejection in liver transplantation is a kind of T cell-mediated inflammatory disease accompanied by inflammatory cell infiltration. However, the effect of acute allograft rejection on the immunological characteristics of TCRs in peripheral blood mononuclear cell is unknown. In this study, we characterized the pattern of the human T cell receptor beta chain (TRB) and immunoglobulin heavy chain (IGH) complementarity-determining region 3 (CDR3) repertoires via high-throughput sequencing in 11 acute allograft rejection (AG) cases, 23 patients with stable allograft liver function (ST) who had liver transplantation performed and 20 healthy controls (HC). The diversity of TRB-CDR3 was significantly reduced in the AG group compared with the ST group and healthy controls (HC). The CDR3 and N-addition length distribution were not significantly different between the AG and ST groups. However, N-addition length distribution was significantly changed compared to HC. It seemed that AG used more short N-additions and healthy people used more long N-additions in TRB-CDR3 repertoire. Our findings suggested that the TRB-CDR3 region of AG had distinctive V gene use compared with that of HC. The characteristics of ST seemed to be in between those of AG and HC although the difference is not significant. Cluster analysis showed that the TRB repertoire could not effectively distinguish AG from ST. This research might give to a better understanding of the immune process of liver transplantation.
Collapse
Affiliation(s)
- Fei-Fei Han
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hua Fan
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lu-Lu Ren
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hua-Guang Wang
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chunlin Wang
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA
| | - Xu Ma
- Department of Genetics, National Research Institute for Family Planning, Beijing, China.,Graduate School of Peking, Union Medical College, Beijing, China
| | - Li-Hong Liu
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qiang He
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chang-Long Guo
- Department of Genetics, National Research Institute for Family Planning, Beijing, China.,Graduate School of Peking, Union Medical College, Beijing, China
| |
Collapse
|
17
|
Buhler S, Bettens F, Dantin C, Ferrari-Lacraz S, Ansari M, Mamez AC, Masouridi-Levrat S, Chalandon Y, Villard J. Genetic T-cell receptor diversity at 1 year following allogeneic hematopoietic stem cell transplantation. Leukemia 2019; 34:1422-1432. [DOI: 10.1038/s41375-019-0654-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/23/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
|
18
|
Yun HD, Varma A, Hussain MJ, Nathan S, Brunstein C. Clinical Relevance of Immunobiology in Umbilical Cord Blood Transplantation. J Clin Med 2019; 8:1968. [PMID: 31739455 PMCID: PMC6912281 DOI: 10.3390/jcm8111968] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/28/2019] [Accepted: 11/07/2019] [Indexed: 12/19/2022] Open
Abstract
Umbilical cord blood transplantation (UCBT) has been an important donor source for allogeneic hematopoietic stem cell transplantation, especially for patients who lack suitable matched donors. UCBT provides unique practical advantages, such as lower risks of graft-versus-host-disease (GVHD), permissive HLA mismatch, and ease of procurement. However, there are clinical challenges in UCBT, including high infection rates and treatment-related mortality in selected patient groups. These clinical advantages and challenges are tightly linked with cell-type specific immune reconstitution (IR). Here, we will review IR, focusing on T and NK cells, and the impact of IR on clinical outcomes. Better understanding of the immune biology in UCBT will allow us to further advance this field with improved clinical practice.
Collapse
Affiliation(s)
- Hyun Don Yun
- Division of Hematology, Oncology and Cellular Therapy, Department of Medicine, Rush University, Chicago, IL 60091, USA; (H.D.Y.); (A.V.); (M.J.H.); (S.N.)
| | - Ankur Varma
- Division of Hematology, Oncology and Cellular Therapy, Department of Medicine, Rush University, Chicago, IL 60091, USA; (H.D.Y.); (A.V.); (M.J.H.); (S.N.)
| | - Mohammad J. Hussain
- Division of Hematology, Oncology and Cellular Therapy, Department of Medicine, Rush University, Chicago, IL 60091, USA; (H.D.Y.); (A.V.); (M.J.H.); (S.N.)
| | - Sunita Nathan
- Division of Hematology, Oncology and Cellular Therapy, Department of Medicine, Rush University, Chicago, IL 60091, USA; (H.D.Y.); (A.V.); (M.J.H.); (S.N.)
| | - Claudio Brunstein
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 60612, USA
| |
Collapse
|
19
|
Kamachi K, Shindo T, Miyahara M, Kitaura K, Akashi M, Shin-I T, Suzuki R, Oshima K, Kimura S. Epstein-Barr virus-related diffuse large B-cell lymphoma in mogamulizumab-treated adult T-cell leukemia with incomplete T-cell reconstitution. Int J Hematol 2018; 109:221-227. [PMID: 30368656 DOI: 10.1007/s12185-018-2552-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 10/20/2018] [Accepted: 10/22/2018] [Indexed: 11/24/2022]
Abstract
Adult T-cell leukemia (ATL) is an aggressive mature T-cell malignancy with a poor prognosis. The anti-C-C motif chemokine receptor 4 (CCR4) antibody mogamulizumab (moga) reduces ATL cells and induces reconstitution of polyclonal T cells; however, ATL cases often remain resistant and moga sometimes causes fatal immunopathology. Epstein-Barr virus (EBV)-related B-cell lymphoma develops in severely immunocompromised subjects, and is particularly associated with impaired T-cell immunity. Here, we report an ATL patient who had received conventional chemotherapy plus moga, and subsequently developed EBV-related diffuse large B-cell lymphoma (DLBCL) of the central nervous system. Next-generation sequencing-based T-cell receptor repertoire analyses identified residual abnormal clones and revealed that reconstitution of polyclonal T cells was incomplete, even after moga treatment. Furthermore, a skin rash that developed after moga treatment was found to contain ATL clones. This case suggests that the limited therapeutic effects of moga and incomplete T-cell reconstitution are associated with severely impaired T-cell immunity and subsequent development of EBV-related DLBCL.
Collapse
Affiliation(s)
- Kazuharu Kamachi
- Department of Internal Medicine, Karatsu Red Cross Hospital, Karatsu, Japan.,Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Takero Shindo
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan. .,Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogo-in, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Masaharu Miyahara
- Department of Internal Medicine, Karatsu Red Cross Hospital, Karatsu, Japan
| | | | - Michiaki Akashi
- Department of Pathology, Karatsu Red Cross Hospital, Karatsu, Japan
| | | | - Ryuji Suzuki
- Repertoire Genesis Inc, Ibaraki, Japan.,Department of Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, Sagamihara, Japan
| | - Koichi Oshima
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
20
|
Ono K, Murata K, Miyazaki A, Tachibana N, Nakamura T, Nishimura R, Yachie A, Kawa K, Shiobara S. Late-onset hemophagocytic lymphohistiocytosis with varicella zoster virus and Epstein-Barr virus co-infection after umbilical cord blood transplantation. Ann Hematol 2018; 97:1493-1495. [PMID: 29568988 DOI: 10.1007/s00277-018-3306-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/12/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Koya Ono
- Department of Hematology, Soka Municipal Hospital, Soka, Japan. .,Department of Hematology and Rheumatology, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Ken Murata
- Department of Hematology, Soka Municipal Hospital, Soka, Japan.,Departments of Hematology, Suwa Red Cross Hospital, Suwa, Japan
| | - Atsushi Miyazaki
- Departments of Dermatology, Suwa Red Cross Hospital, Suwa, Japan
| | - Naoki Tachibana
- Departments of Nephrology, Suwa Red Cross Hospital, Suwa, Japan
| | | | | | - Akihiro Yachie
- Department of Pediatrics, Kanazawa University, Kanazawa, Japan
| | - Keisei Kawa
- Division of Hematology/Oncology, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Shintaro Shiobara
- Departments of Hematology, Suwa Red Cross Hospital, Suwa, Japan.,Japanese Red Cross Ishikawa Blood Center, Kanazawa, Japan
| |
Collapse
|
21
|
Impaired thymopoiesis predicts for a high risk of severe infections after reduced intensity conditioning without anti-thymocyte globulin in double umbilical cord blood transplantation. Bone Marrow Transplant 2018; 53:673-682. [DOI: 10.1038/s41409-018-0103-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 01/06/2023]
|
22
|
Solders M, Erkers T, Gorchs L, Poiret T, Remberger M, Magalhaes I, Kaipe H. Mucosal-Associated Invariant T Cells Display a Poor Reconstitution and Altered Phenotype after Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2017; 8:1861. [PMID: 29312341 PMCID: PMC5742569 DOI: 10.3389/fimmu.2017.01861] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/07/2017] [Indexed: 12/21/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are innate-like T cells which are important in the defense against certain bacteria and yeast. The reconstitution of MAIT cells after allogeneic hematopoietic stem cell transplantation (HSCT) is not known. We investigated MAIT cell phenotype and function in 17 patients devoid of relapse and severe graft-versus-host disease (GvHD) in paired samples collected 1-2, 3-6, 12, and 24 months after transplantation. Data were compared to 17 healthy controls (HC), as well as 22 patients with acute GvHD grade 2-3. The frequency of MAIT cells within CD3+ cells was approximately 10-fold lower than in HC and did not increase over the 2 years following HSCT. MAIT cells in HSCT patients displayed an elevated expression of CD69 and intracellular granzyme B and were predominantly composed of CD4/CD8 double-negative cells. The expression of PD-1 on MAIT cells was low and did not change during the observational time, whereas the CD3+CD161dim/negTCRVα7.2dim/neg cells (non-MAIT T cells) displayed a high expression early after HSCT that decreased to normal levels at 24 months. MAIT cells collected 2-6 months post-HSCT showed an impaired IFN-γ and perforin response after bacterial stimulation, but the response was restored at 24 months. Patients with acute GvHD had similar proportions of MAIT cells as patients with grade 0-1, but consisted mainly of CD8+ cells. Finally, MAIT cells were more sensitive to cyclosporine A and sirolimus than non-MAIT T cells. To conclude, MAIT cell reconstitution following HSCT is deficient compared to non-MAIT T cells and GvHD grade ≥2 is not correlated with MAIT cell frequency. MAIT cell functionality was impaired early after HSCT, but restored at 24 months post-HSCT. MAIT cells have an increased sensibility to common immunosuppressive drugs, which maybe could explain their hampered reconstitution after HSCT.
Collapse
Affiliation(s)
- Martin Solders
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tom Erkers
- Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, United States
| | - Laia Gorchs
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Poiret
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Helen Kaipe
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
23
|
Shindo T, Kitaura K, Ureshino H, Kamachi K, Miyahara M, Doi K, Watanabe T, Sueoka E, Shin-I T, Suzuki R, Kimura S. Deep sequencing of the T cell receptor visualizes reconstitution of T cell immunity in mogamulizumab-treated adult T cell leukemia. Oncoimmunology 2017; 7:e1405204. [PMID: 29399406 DOI: 10.1080/2162402x.2017.1405204] [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: 06/23/2017] [Revised: 10/26/2017] [Accepted: 11/09/2017] [Indexed: 10/18/2022] Open
Abstract
Although the anti-CCR4 antibody mogamulizumab (moga) shows striking antitumor activity against adult T cell leukemia (ATL), it can also cause fatal immunological pathology such as severe skin rash and graft-versus-host disease, which might be attributed to depletion of CCR4+ regulatory T cells. We previously showed that next generation sequencing enables precise analysis of the T cell receptor (TCR) repertoire, and we here used the technique to reveal the immunological dynamics in moga-treated ATL patients. Treatment with moga resulted in remarkable reduction or elimination of clonal cells, and enhanced reconstitution of non-tumor polyclonal CD4+ T cells and oligoclonal CD8+ T cells. Interestingly, cutaneous T cells infiltrating moga-related skin rashes did not share the same major clones in peripheral blood, which minimizes the possibility of cross-reaction. Thus, deep sequencing of the TCR can reveal the immune reconstitution of moga-treated ATL and provides powerful insights into its mode of action.
Collapse
Affiliation(s)
- Takero Shindo
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan.,Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Hiroshi Ureshino
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| | - Kazuharu Kamachi
- Department of Internal Medicine, Karatsu Red Cross Hospital, Karatsu, Japan
| | - Masaharu Miyahara
- Department of Internal Medicine, Karatsu Red Cross Hospital, Karatsu, Japan
| | - Kazuko Doi
- Department of Dermatology, Karatsu Red Cross Hospital, Karatsu, Japan
| | - Tatsuro Watanabe
- Department of Laboratory Medicine, Saga University Hospital, Saga, Japan
| | - Eisaburo Sueoka
- Department of Laboratory Medicine, Saga University Hospital, Saga, Japan.,Department of Clinical Laboratory Medicine, Saga University School of Medicine, Saga, Japan
| | | | - Ryuji Suzuki
- Repertoire Genesis, Inc., Ibaraki, Japan.,Department of Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, Sagamihara, Japan
| | - Shinya Kimura
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| |
Collapse
|
24
|
Umbilical Cord Blood Cytomegalovirus Serostatus Does Not Have an Impact on Outcomes of Umbilical Cord Blood Transplantation for Acute Leukemia. Biol Blood Marrow Transplant 2017; 23:1729-1735. [DOI: 10.1016/j.bbmt.2017.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/28/2017] [Indexed: 01/27/2023]
|
25
|
de Koning C, Admiraal R, Nierkens S, Boelens JJ. Immune reconstitution and outcomes after conditioning with anti-thymocyte-globulin in unrelated cord blood transplantation; the good, the bad, and the ugly. Stem Cell Investig 2017; 4:38. [PMID: 28607912 DOI: 10.21037/sci.2017.05.02] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/17/2017] [Indexed: 01/07/2023]
Abstract
Unrelated umbilical cord blood transplantation (UCBT) exhibits a low risk of graft-versus-host-disease (GvHD) and has unique potent anti-virus and anti-leukemia effects. Anti-thymocyte globulin (ATG) in the conditioning regimen for UCBT is successful in reducing graft rejection and GvHD. Nevertheless, this beneficial effect of ATG coincides with its detrimental effect on immune reconstitution. The latter directly relates to a high incidence of viral infections and leukemia relapses. ATG has been used in transplant patients for over 30 years. In recent years, the knowledge on the mechanisms of action of ATG and its implementation in the UCBT setting has increased dramatically. Important data became available showing the highly variable pharmacokinetics (PK) of ATG and its consequence on outcome measures. Here, we review the effects of ATG on immune reconstitution and subsequent outcomes after UCBT, and describe the mechanisms causing these effects. We highlight the importance of optimizing ATG exposure before and after UCBT and discuss strategies to maintain the 'good' and overcome the 'bad and ugly' effects of ATG on UCBT outcome.
Collapse
Affiliation(s)
- Coco de Koning
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rick Admiraal
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.,Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jaap Jan Boelens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.,Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
26
|
Gaballa A, Sundin M, Stikvoort A, Abumaree M, Uzunel M, Sairafi D, Uhlin M. T Cell Receptor Excision Circle (TREC) Monitoring after Allogeneic Stem Cell Transplantation; a Predictive Marker for Complications and Clinical Outcome. Int J Mol Sci 2016; 17:E1705. [PMID: 27727179 PMCID: PMC5085737 DOI: 10.3390/ijms17101705] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/26/2016] [Accepted: 09/29/2016] [Indexed: 12/22/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a well-established treatment modality for a variety of malignant diseases as well as for inborn errors of the metabolism or immune system. Regardless of disease origin, good clinical effects are dependent on proper immune reconstitution. T cells are responsible for both the beneficial graft-versus-leukemia (GVL) effect against malignant cells and protection against infections. The immune recovery of T cells relies initially on peripheral expansion of mature cells from the graft and later on the differentiation and maturation from donor-derived hematopoietic stem cells. The formation of new T cells occurs in the thymus and as a byproduct, T cell receptor excision circles (TRECs) are released upon rearrangement of the T cell receptor. Detection of TRECs by PCR is a reliable method for estimating the amount of newly formed T cells in the circulation and, indirectly, for estimating thymic function. Here, we discuss the role of TREC analysis in the prediction of clinical outcome after allogeneic HSCT. Due to the pivotal role of T cell reconstitution we propose that TREC analysis should be included as a key indicator in the post-HSCT follow-up.
Collapse
Affiliation(s)
- Ahmed Gaballa
- Department of Oncology and Pathology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| | - Mikael Sundin
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
- Pediatric Blood Disorders, Immunodeficiency and Stem Cell Transplantation, Astrid Lindgren Children's Hospital, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| | - Arwen Stikvoort
- Department of Oncology and Pathology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| | - Muhamed Abumaree
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, KSA-11461 Riyadh, Saudi Arabia.
| | - Mehmet Uzunel
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| | - Darius Sairafi
- Department of Oncology and Pathology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| | - Michael Uhlin
- Department of Oncology and Pathology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
27
|
Guo C, Wang Q, Cao X, Yang Y, Liu X, An L, Cai R, Du M, Wang G, Qiu Y, Peng Z, Han J, Ni S, Tan X, Jin L, Yu S, Wang H, Wang C, Wang X, Ma X. High-Throughput Sequencing Reveals Immunological Characteristics of the TRB-/IgH-CDR3 Region of Umbilical Cord Blood. J Pediatr 2016; 176:69-78.e1. [PMID: 27373756 DOI: 10.1016/j.jpeds.2016.05.078] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/11/2016] [Accepted: 05/26/2016] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To compare the differences of immunological characteristics between newborn and adults, we performed high-throughput sequencing to reveal the diversity of umbilical cord blood and adult peripheral blood at both T-cell receptor beta chain (TRB) and immunoglobulin heavy chain (IGH) levels. STUDY DESIGN High-throughput sequencing was performed to analyze the expression of TRB-CDR3 and IGH-CDR3 in circulating T and B cells isolated from 20 healthy adults, 56 pregnant women, and 40 newborns. RESULTS Our results revealed different immunological characteristics between newborn and adults, such as distinctive complementarity determining region 3 (CDR3) lengths, usage bias of variable and joining segments, random nucleotide addition, a large number of unique CDR3 peptides, and a greater repertoire diversity. Moreover, each newborn had a distinctive TRB-/IGH-CDR3 repertoire that was independent of the maternal immune status. CONCLUSIONS This study presents comprehensive, unrestricted profiles of the TRB/IGH-CDR3 repertoire of newborns, pregnant women, and healthy adults at a sequence-level resolution. Our data may contribute to a better understanding of the immune system of newborns and benefit the efficient application of umbilical cord blood transplantation in future.
Collapse
MESH Headings
- Adult
- Complementarity Determining Regions/blood
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/immunology
- Female
- Fetal Blood
- High-Throughput Nucleotide Sequencing
- Humans
- Immunoglobulin Heavy Chains/blood
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Heavy Chains/immunology
- Infant, Newborn
- Pregnancy
- Receptors, Antigen, T-Cell, alpha-beta/blood
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- Changlong Guo
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Qidi Wang
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Xiaofang Cao
- Department of Genetics, National Research Institute for Family Planning, Beijing, China.
| | - Ying Yang
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Xin Liu
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Laboratory of Human Genetics, Beijing Hypertension League Institute, Beijing, China
| | - Lisha An
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Ruikun Cai
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Meng Du
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Guangyu Wang
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Yue Qiu
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Zuoqi Peng
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Jian Han
- HudsonAlpha Institute for Biotechnology, Huntsville, AL
| | - Shuhua Ni
- First Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong, China
| | - Xuerui Tan
- First Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong, China
| | - Li Jin
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Song Yu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Healthy Care Hospital, Beijing, China
| | - Huiying Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chunlin Wang
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Stanford Genome Technology Center, Stanford University, Palo Alto, CA
| | - Xingyu Wang
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Laboratory of Human Genetics, Beijing Hypertension League Institute, Beijing, China
| | - Xu Ma
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
28
|
Arruda LCM, de Azevedo JTC, de Oliveira GLV, Scortegagna GT, Rodrigues ES, Palma PVB, Brum DG, Guerreiro CT, Marques VD, Barreira AA, Covas DT, Simões BP, Voltarelli JC, Oliveira MC, Malmegrim KCR. Immunological correlates of favorable long-term clinical outcome in multiple sclerosis patients after autologous hematopoietic stem cell transplantation. Clin Immunol 2016; 169:47-57. [PMID: 27318116 DOI: 10.1016/j.clim.2016.06.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/13/2016] [Accepted: 06/13/2016] [Indexed: 12/29/2022]
Abstract
High dose immunosuppression followed by autologous hematopoietic stem cell transplantation (AHSCT) induces prolonged clinical remission in multiple sclerosis (MS) patients. However, how patient immune profiles are associated with clinical outcomes has not yet been completely elucidated. In this study, 37 MS patients were assessed for neurological outcomes, thymic function and long-term immune reconstitution after AHSCT. Patients were followed for a mean (SD) of 68.5 (13.9) months post-transplantation and were retrospectively clustered into progression- and non-progression groups, based on Expanded Disease Status Scale (EDSS) outcomes at last visit. After AHSCT, both patient groups presented increased regulatory T-cell subset counts, early expansion of central- and effector-memory CD8(+)T-cells and late thymic reactivation. However, the non-progression group presented early expansion of PD-1(+)CD8(+)T-cells and of PD-1-expressing CD19(+) B-cells. Here, we suggest that along with increased numbers of regulatory T-cell subsets, PD-1 inhibitory signaling is one possible immunoregulatory mechanism by which AHSCT restores immune tolerance in MS patients.
Collapse
Affiliation(s)
- Lucas C M Arruda
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Júlia T C de Azevedo
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gislane L V de Oliveira
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gabriela T Scortegagna
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Evandra S Rodrigues
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Patrícia V B Palma
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Doralina G Brum
- Department of Neuroscience and Behavioral Science, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Neurology, Psychology and Psychiatry, School of Medicine of Botucatu, Universidade Estadual Paulista, UNESP, Botucatu, Brazil
| | - Carlos T Guerreiro
- Department of Neuroscience and Behavioral Science, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa D Marques
- Department of Neuroscience and Behavioral Science, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Amilton A Barreira
- Department of Neuroscience and Behavioral Science, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas T Covas
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Belinda P Simões
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Júlio C Voltarelli
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Carolina Oliveira
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen C R Malmegrim
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
29
|
TNF-alpha and Notch signaling regulates the expression of HOXB4 and GATA3 during early T lymphopoiesis. In Vitro Cell Dev Biol Anim 2016; 52:920-934. [PMID: 27251160 DOI: 10.1007/s11626-016-0055-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
Abstract
During the early thymus colonization, Notch signaling activation on hematopoietic progenitor cells (HPCs) drives proliferation and T cell commitment. Although these processes are driven by transcription factors such as HOXB4 and GATA3, there is no evidence that Notch directly regulates their transcription. To evaluate the role of NOTCH and TNF signaling in this process, human CD34+ HPCs were cocultured with OP9-DL1 cells, in the presence or absence of TNF. The use of a Notch signaling inhibitor and a protein synthesis inhibitor allowed us to distinguish primary effects, mediated by direct signaling downstream Notch and TNF, from secondary effects, mediated by de novo synthesized proteins. A low and physiologically relevant concentration of TNF promoted T lymphopoiesis in OP9-DL1 cocultures. TNF positively modulated the expression of both transcripts in a Notch-dependent manner; however, GATA3 induction was mediated by a direct mechanism, while HOXB4 induction was indirect. Induction of both transcripts was repressed by a GSK3β inhibitor, indicating that activation of canonical Wnt signaling inhibits rather than induces their expression. Our study provides novel evidences of the mechanisms integrating Notch and TNF-alpha signaling in the transcriptional induction of GATA3 and HOXB4. This mechanism has direct implications in the control of self-renewal, proliferation, commitment, and T cell differentiation.
Collapse
|
30
|
Lucchini G, Perales MA, Veys P. Immune reconstitution after cord blood transplantation: peculiarities, clinical implications and management strategies. Cytotherapy 2016; 17:711-722. [PMID: 25946726 DOI: 10.1016/j.jcyt.2015.03.614] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/12/2015] [Indexed: 12/26/2022]
Abstract
Umbilical cord blood (UCB) is now widely used as an alternative hematopoietic stem cell source for patients lacking closely matched related or unrelated adult donors. UCB transplantation has traditionally been associated with delayed engraftment, poor immune reconstitution and consequent increased risk of infection. More recent clinical studies, however, suggest that conditioning regimens and in particular the omission of in vivo T-cell depletion may play a crucial role in post-transplant T-cell expansion, facilitating a uniquely rapid immune recovery after UCB transplantation. The peculiar characteristics of UCB cells, the importance of thymic function and the role of conditioning regimens and graft-versus-host disease influencing immune reconstitution are described. The last part of the review reports available data on UCB, as well as third-party peripheral blood derived anti-viral cell therapy, which provides a novel approach to rescue UCB recipients with viral complications in the post-transplant period.
Collapse
Affiliation(s)
- Giovanna Lucchini
- Bone Marrow Transplantation Department, Great Ormond Street Hospital, London, United Kingdom.
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, USA
| | - Paul Veys
- Bone Marrow Transplantation Department, Great Ormond Street Hospital, London, United Kingdom
| |
Collapse
|
31
|
Alanio C, Nicoli F, Sultanik P, Flecken T, Perot B, Duffy D, Bianchi E, Lim A, Clave E, van Buuren MM, Schnuriger A, Johnsson K, Boussier J, Garbarg-Chenon A, Bousquet L, Mottez E, Schumacher TN, Toubert A, Appay V, Heshmati F, Thimme R, Pol S, Mallet V, Albert ML. Bystander hyperactivation of preimmune CD8+ T cells in chronic HCV patients. eLife 2015; 4. [PMID: 26568315 PMCID: PMC4752008 DOI: 10.7554/elife.07916] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 11/12/2015] [Indexed: 01/21/2023] Open
Abstract
Chronic infection perturbs immune homeostasis. While prior studies have reported dysregulation of effector and memory cells, little is known about the effects on naïve T cell populations. We performed a cross-sectional study of chronic hepatitis C (cHCV) patients using tetramer-associated magnetic enrichment to study antigen-specific inexperienced CD8(+) T cells (i.e., tumor or unrelated virus-specific populations in tumor-free and sero-negative individuals). cHCV showed normal precursor frequencies, but increased proportions of memory-phenotype inexperienced cells, as compared to healthy donors or cured HCV patients. These observations could be explained by low surface expression of CD5, a negative regulator of TCR signaling. Accordingly, we demonstrated TCR hyperactivation and generation of potent CD8(+) T cell responses from the altered T cell repertoire of cHCV patients. In sum, we provide the first evidence that naïve CD8(+) T cells are dysregulated during cHCV infection, and establish a new mechanism of immune perturbation secondary to chronic infection.
Collapse
Affiliation(s)
- Cécile Alanio
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Centre d'Immunologie Humaine, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | - Francesco Nicoli
- Sorbonne Universités, UPMC Univ Paris 06, DNU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,Emory, , United States
| | - Philippe Sultanik
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | - Tobias Flecken
- The University Medical Center Freiburg, Department of Internal Medicine II, Albert-Ludwigs-Universität, Freiberg, Germany
| | - Brieuc Perot
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | - Darragh Duffy
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Centre d'Immunologie Humaine, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | | | - Annick Lim
- Plateforme d'Immunoscope, Institut Pasteur, Paris, France
| | - Emmanuel Clave
- Hôpital Saint-Louis, Assistance publique - hôpitaux de Paris, Paris, France
| | - Marit M van Buuren
- Department of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Aurélie Schnuriger
- Laboratoire de virologie, Hôpital Armand-Trousseau, Assistance publique - hôpitaux de Paris, Paris, France
| | - Kerstin Johnsson
- Mathematics, Faculty of Engineering, Lunds University, Lund, Sweden
| | - Jeremy Boussier
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Centre d'Immunologie Humaine, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | - Antoine Garbarg-Chenon
- Laboratoire de virologie, Hôpital Armand-Trousseau, Assistance publique - hôpitaux de Paris, Paris, France
| | | | - Estelle Mottez
- Centre d'Immunologie Humaine, Institut Pasteur, Paris, France
| | - Ton N Schumacher
- Department of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Antoine Toubert
- Hôpital Saint-Louis, Assistance publique - hôpitaux de Paris, Paris, France
| | - Victor Appay
- Sorbonne Universités, UPMC Univ Paris 06, DNU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,Emory, , United States
| | | | - Robert Thimme
- The University Medical Center Freiburg, Department of Internal Medicine II, Albert-Ludwigs-Universität, Freiberg, Germany
| | | | | | - Matthew L Albert
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Centre d'Immunologie Humaine, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| |
Collapse
|
32
|
Development of a diverse human T-cell repertoire despite stringent restriction of hematopoietic clonality in the thymus. Proc Natl Acad Sci U S A 2015; 112:E6020-7. [PMID: 26483497 DOI: 10.1073/pnas.1519118112] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The fate and numbers of hematopoietic stem cells (HSC) and their progeny that seed the thymus constitute a fundamental question with important clinical implications. HSC transplantation is often complicated by limited T-cell reconstitution, especially when HSC from umbilical cord blood are used. Attempts to improve immune reconstitution have until now been unsuccessful, underscoring the need for better insight into thymic reconstitution. Here we made use of the NOD-SCID-IL-2Rγ(-/-) xenograft model and lentiviral cellular barcoding of human HSCs to study T-cell development in the thymus at a clonal level. Barcoded HSCs showed robust (>80% human chimerism) and reproducible myeloid and lymphoid engraftment, with T cells arising 12 wk after transplantation. A very limited number of HSC clones (<10) repopulated the xenografted thymus, with further restriction of the number of clones during subsequent development. Nevertheless, T-cell receptor rearrangements were polyclonal and showed a diverse repertoire, demonstrating that a multitude of T-lymphocyte clones can develop from a single HSC clone. Our data imply that intrathymic clonal fitness is important during T-cell development. As a consequence, immune incompetence after HSC transplantation is not related to the transplantation of limited numbers of HSC but to intrathymic events.
Collapse
|
33
|
Li Y, Xu L. Evaluation of TCR repertoire diversity in patients after hematopoietic stem cell transplantation. Stem Cell Investig 2015; 2:17. [PMID: 27358885 DOI: 10.3978/j.issn.2306-9759.2015.09.01] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 08/19/2015] [Indexed: 12/16/2022]
Abstract
T-cell receptor (TCR) repertoire analyses have been widely used to identify T cell populations of interest in cancer and autoimmunity and for characterizing immune repertoire reconstitution after hematopoietic stem cell transplantation (HSCT). Several decades of development and progress have led to the use of techniques for evaluating TCR repertoires in a more comprehensive, unbiased and fast manner, and the mechanisms of T cell immune reconstitution after HSCT and the new approaches used for recovering T cell repertoire diversity post HSCT have been more exhaustively documented to some degree. To better understand and characterize this progress, here we review recent studies on TCR repertoire diversity recovery in patients with leukemia and autoimmune disease who have received HSCT, impact factors and improvements in approaches for TCR repertoire recovery after HSCT.
Collapse
Affiliation(s)
- Yangqiu Li
- 1 Institute of Hematology, Medical College, 2 Department of Hematology, First Affiliated Hospital, 3 Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Ling Xu
- 1 Institute of Hematology, Medical College, 2 Department of Hematology, First Affiliated Hospital, 3 Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou 510632, China
| |
Collapse
|
34
|
de Koning C, Plantinga M, Besseling P, Boelens JJ, Nierkens S. Immune Reconstitution after Allogeneic Hematopoietic Cell Transplantation in Children. Biol Blood Marrow Transplant 2015; 22:195-206. [PMID: 26341398 DOI: 10.1016/j.bbmt.2015.08.028] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/25/2015] [Indexed: 12/14/2022]
Abstract
Allogeneic (allo) hematopoietic cell transplantation (HCT) has evolved into a potent curative treatment option for a variety of malignant and nonmalignant diseases. The occurrence of complications and mortality after allo-HCT is, however, still high and is strongly associated with immune reconstitution (IR). Therefore, detailed information on IR through immunomonitoring is crucial to improve survival chances after HCT. To date, information about the reconstituting immune system after allo-HCT in pediatric patients is mostly derived from routine standard-of-care measurements. More profound knowledge on IR may provide tools to better predict and modulate adverse reactions and, subsequently, improve survival chances. Here, we provide an overview of IR (eg, immune cell subsets and circulating chemokines/cytokines) after allo-HCT in children, taking into account different cell sources and serotherapy, and discuss strategies to enhance immunomonitoring. We conclude that available IR data after allo-HCT contain limited information on immune cell families (mostly only generic T, B, and NK cells), which would improve with more detailed information on reconstituting cell subsets or effector cell functionality at earlier time points (<1 month). In addition, secretome data (eg, multiplex cytokine/chemokine profiles) could add to the understanding of IR mechanisms and cell functionality and may even provide (early) biomarkers for individual disease outcome, such as viral reactivity, graft-versus-host disease, or graft-versus-leukemia. The present data and suggestions for more detailed, standardized, and harmonized immunomonitoring in future (pediatric) allo-HCT studies will pave the path to "precision transplantation:" an individualized HCT approach (including conditioning), based on detailed information on IR and biomarkers, aiming to reduce transplantation related mortality and relapse, and subsequently improve survival chances.
Collapse
Affiliation(s)
- Coco de Koning
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Maud Plantinga
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Paul Besseling
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jaap Jan Boelens
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands; Pediatric Blood and Marrow Transplantation Program, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
35
|
The role of the thymus in T-cell immune reconstitution after umbilical cord blood transplantation. Blood 2014; 124:3201-11. [PMID: 25287708 DOI: 10.1182/blood-2014-07-589176] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Umbilical cord blood (UCB) is an alternative source of hematopoietic stem cells for patients without HLA-matched adult donors. UCB contains a low number of nucleated cells and mostly naive T cells, resulting in prolonged time to engraftment and lack of transferred T-cell memory. Although the first phase of T-cell reconstitution after UCB transplantation (UCBT) depends on peripheral expansion of transferred T cells, permanent T-cell reconstitution is mediated via a central mechanism, which depends on de novo production of naive T lymphocytes by the recipient's thymus from donor-derived lymphoid-myeloid progenitors (LMPs). Thymopoiesis can be assessed by quantification of recent thymic emigrants, T-cell receptor excision circle levels, and T-cell receptor repertoire diversity. These assays are valuable tools for monitoring posttransplantation thymic recovery, but more importantly they have shown the significant prognostic value of thymic reconstitution for clinical outcomes after UCBT, including opportunistic infections, disease relapse, and overall survival. Strategies to improve thymic entry and differentiation of LMPs and to accelerate recovery of the thymic stromal microenvironment may improve thymic lymphopoiesis. Here, we discuss the mechanisms and clinical implications of thymic recovery and new approaches to improve reconstitution of the T-cell repertoire after UCBT.
Collapse
|
36
|
Comparative analysis of unrelated cord blood transplantation and HLA-matched sibling hematopoietic stem cell transplantation in children with high-risk or advanced acute leukemia. Ann Hematol 2014; 94:473-80. [DOI: 10.1007/s00277-014-2213-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 09/02/2014] [Indexed: 10/24/2022]
|
37
|
Hamadani M, Vos JA, Craig MD. Reply to S. Fuji et al. J Clin Oncol 2014; 32:1860-1. [PMID: 24799471 DOI: 10.1200/jco.2014.55.4048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
38
|
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
|
39
|
Danby R, Rocha V. Improving engraftment and immune reconstitution in umbilical cord blood transplantation. Front Immunol 2014; 5:68. [PMID: 24605111 PMCID: PMC3932655 DOI: 10.3389/fimmu.2014.00068] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/07/2014] [Indexed: 12/31/2022] Open
Abstract
Umbilical cord blood (UCB) is an important source of hematopoietic stem cells (HSC) for allogeneic transplantation when HLA-matched sibling and unrelated donors (MUD) are unavailable. Although the overall survival results for UCB transplantation are comparable to the results with MUD, UCB transplants are associated with slow engraftment, delayed immune reconstitution, and increased opportunistic infections. While this may be a consequence of the lower cell dose in UCB grafts, it also reflects the relative immaturity of cord blood. Furthermore, limited cell numbers and the non-availability of donor lymphocyte infusions currently prevent the use of post-transplant cellular immunotherapy to boost donor-derived immunity to treat infections, mixed chimerism, and disease relapse. To further develop UCB transplantation, many strategies to enhance engraftment and immune reconstitution are currently under investigation. This review summarizes our current understanding of engraftment and immune recovery following UCB transplantation and why this differs from allogeneic transplants using other sources of HSC. It also provides a comprehensive overview of promising techniques being used to improve myeloid and lymphoid recovery, including expansion, homing, and delivery of UCB HSC; combined use of UCB with third-party donors; isolation and expansion of natural killer cells, pathogen-specific T cells, and regulatory T cells; methods to protect and/or improve thymopoiesis. As many of these strategies are now in clinical trials, it is anticipated that UCB transplantation will continue to advance, further expanding our understanding of UCB biology and HSC transplantation.
Collapse
Affiliation(s)
- Robert Danby
- Department of Haematology, Churchill Hospital, Oxford University Hospitals NHS Trust , Oxford , UK ; NHS Blood and Transplant, John Radcliffe Hospital , Oxford , UK ; Eurocord, Hôpital Saint Louis APHP, University Paris VII IUH , Paris , France
| | - Vanderson Rocha
- Department of Haematology, Churchill Hospital, Oxford University Hospitals NHS Trust , Oxford , UK ; NHS Blood and Transplant, John Radcliffe Hospital , Oxford , UK ; Eurocord, Hôpital Saint Louis APHP, University Paris VII IUH , Paris , France
| |
Collapse
|
40
|
Hakim FT, Gress RE. Immunosenescence: immune deficits in the elderly and therapeutic strategies to enhance immune competence. Expert Rev Clin Immunol 2014; 1:443-58. [DOI: 10.1586/1744666x.1.3.443] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
41
|
Servais S, Lengline E, Porcher R, Carmagnat M, Peffault de Latour R, Robin M, Sicre de Fontebrune F, Clave E, Maki G, Granier C, Xhaard A, Dhedin N, Molina JM, Toubert A, Moins-Teisserenc H, Socie G. Long-term immune reconstitution and infection burden after mismatched hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2014; 20:507-17. [PMID: 24406505 DOI: 10.1016/j.bbmt.2014.01.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 01/02/2014] [Indexed: 12/19/2022]
Abstract
Mismatched unrelated donor (MMUD) or umbilical cord blood (UCB) can be chosen as alternative donors for allogeneic stem cell transplantation but might be associated with long-lasting immune deficiency. Sixty-six patients who underwent a first transplantation from either UCB (n = 30) or 9/10 MMUD (n = 36) and who survived beyond 3 months were evaluated. Immune reconstitution was prospectively assessed at sequential time points after transplantation. NK, B, CD4(+), and CD8(+) T cells and their naïve and memory subsets, as well as regulatory T cells (Treg), were studied. Detailed analyses on infections occurring after 3 months were also assessed. The 18-month cumulative incidences of infection-related death were 8% and 3%, and of infections were 72% and 57% after MMUD and UCB transplantation, respectively. Rates of infection per 12 patient-month were roughly 2 overall (1 for bacterial, .9 for viral, and .3 for fungal infections). Memory, naïve CD4(+) and CD8(+)T cells, naïve B cells, and Treg cells reconstitution between the 2 sources were roughly similar. Absolute CD4(+)T cells hardly reached 500 per μL by 1 year after transplantation and most B cells were of naïve phenotype. Correlations between immune reconstitution and infection were then performed by multivariate analyses. Low CD4(+) and high CD8(+)T cells absolute counts at 3 months were linked to increased risks of overall and viral (but not bacterial) infections. When assessing for the naïve/memory phenotypes at 3 months among the CD4(+) T cell compartment, higher percentages of memory subsets were protective against late infections. Central memory CD4(+)T cells protected against overall and bacterial infections; late effector memory CD4(+)T cells protected against overall, bacterial, and viral infections. To the contrary, high percentage of effector- and late effector-memory subsets at 3 months among the CD8(+) T cell compartment predicted higher risks for viral infections. Patients who underwent transplantation from alternative donors represent a population with very high risk of infection. Detailed phenotypic analysis of immune reconstitution may help to evaluate infection risk and to adjust infection prophylaxis.
Collapse
Affiliation(s)
- Sophie Servais
- Service d'Hématologie Greffe, AP-HP Hôpital Saint Louis, Paris, France
| | - Etienne Lengline
- Service d'Hématologie Greffe, AP-HP Hôpital Saint Louis, Paris, France
| | | | | | | | - Marie Robin
- Service d'Hématologie Greffe, AP-HP Hôpital Saint Louis, Paris, France
| | | | - Emmanuel Clave
- Immunologie, AP-HP Hôpital Saint Louis, Paris, France; Inserm U 940, Hôpital Saint Louis, Paris, France
| | - Guitta Maki
- Immunologie, AP-HP Hôpital Saint Louis, Paris, France
| | | | - Alienor Xhaard
- Service d'Hématologie Greffe, AP-HP Hôpital Saint Louis, Paris, France
| | - Nathalie Dhedin
- Service d'Hématologie Greffe, AP-HP Hôpital Saint Louis, Paris, France
| | - Jean-Michel Molina
- Service des Maladies Infectieuses et Tropicales, AP-HP Hôpital Saint Louis, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, France
| | - Antoine Toubert
- Immunologie, AP-HP Hôpital Saint Louis, Paris, France; Inserm U 940, Hôpital Saint Louis, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, France
| | - Hélène Moins-Teisserenc
- Immunologie, AP-HP Hôpital Saint Louis, Paris, France; Inserm U 940, Hôpital Saint Louis, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, France
| | - Gérard Socie
- Service d'Hématologie Greffe, AP-HP Hôpital Saint Louis, Paris, France; Inserm U 940, Hôpital Saint Louis, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, France.
| |
Collapse
|
42
|
Lane JP, Evans PTG, Nademi Z, Barge D, Jackson A, Hambleton S, Flood TJ, Cant AJ, Abinun M, Slatter MA, Gennery AR. Low-dose serotherapy improves early immune reconstitution after cord blood transplantation for primary immunodeficiencies. Biol Blood Marrow Transplant 2013; 20:243-9. [PMID: 24225641 PMCID: PMC7110834 DOI: 10.1016/j.bbmt.2013.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 11/06/2013] [Indexed: 11/27/2022]
Abstract
Cord blood transplantation (CBT) is curative for many primary immunodeficiencies (PIDs) but is associated with risks of viral infection and graft-versus-host disease (GvHD). Serotherapy reduces GvHD but potentially increases the risk of viral infection by delaying immune reconstitution. Because many PID patients have pre-existing viral infections, the optimal dose of serotherapy is unclear. We performed a retrospective analysis in 34 consecutive PID patients undergoing CBT and compared immune reconstitution, viral infection, GvHD, mortality, and long-term immune function between high-dose (n = 11) and low-dose (n = 9) serotherapy. Serotherapy dose had no effect on neutrophil engraftment. Median CD3+ engraftment occurred at 92.5 and 97 days for high- and low-dose serotherapy, respectively. The low-dose serotherapy group had higher CD3+, CD4+, and early thymic emigrant counts at 4 months compared with the high-dose group. GvHD severity and number of viral infections did not differ between serotherapy doses. Survival from the transplantation process was 90.9% for high-dose and 100% for low-dose groups. In conclusion, low-dose serotherapy enhanced T cell reconstitution and thymopoiesis during the first year after CBT with no increase in GvHD.
Collapse
Affiliation(s)
- Jonathan P Lane
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Philippa T G Evans
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Zohreh Nademi
- Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Dawn Barge
- Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Anthony Jackson
- International Centre for Life, Newcastle upon Tyne, United Kingdom
| | - Sophie Hambleton
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Terry J Flood
- Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Andrew J Cant
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Mario Abinun
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Mary A Slatter
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Andrew R Gennery
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
43
|
Impact of thymoglobulin prior to pediatric unrelated umbilical cord blood transplantation on immune reconstitution and clinical outcome. Blood 2013; 123:126-32. [PMID: 24184682 DOI: 10.1182/blood-2013-05-502385] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In vivo T-cell depletion might contribute to the delayed immune reconstitution observed after unrelated umbilical cord blood transplantation (UCBT). We studied the impact of early, late, and no antithymocyte globulin (ATG) on immune reconstitution and outcome. One hundred twenty seven children receiving UCBT in London or Utrecht were divided into 3 groups: early ATG (days -9 to -5; n = 33), late ATG (days -5 to 0; n = 48), and no ATG (n = 46). The no-ATG group received mycophenolate mofetile + cyclosporin A as graft-versus-host disease (GVHD) prophylaxis, while the ATG groups received cyclosporin A + prednisone. End points studied were survival, immune recovery, infections, and GVHD. The probability of survival was similar in all groups: no ATG, 71% ± 8%; early ATG, 68% ± 9%; and late ATG, 61% ± 7%. CD3(+), CD4(+), and CD4(+)-naive T-cell counts were significantly higher (P < .001) in the no-ATG group at 1, 2, 3, 6, and 12 months post-UCBT. In the no-ATG group, significantly fewer viral reactivations (P = .021) were noted. A higher probability of severe acute GVHD (aGVHD; 31%) was found in the no-ATG group compared with 18% (P = .018) for early-ATG and 5% (P < .001) for late-ATG groups. This was not associated with more chronic GVHD (cGVHD).
Collapse
|
44
|
O'Keefe CL, Risitano AM, Maciejewski JP. Clinical Implications of T Cell Receptor Repertoire Analysis after Allogeneic Stem Cell Transplantation. Hematology 2013; 9:189-98. [PMID: 15204100 DOI: 10.1080/10245330410001701530] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Stem cell transplantation (SCT) constitutes a major challenge to the immune system. Long-term impairment of immunity against various common infectious stimuli leads to increased susceptibility to infectious diseases; in contrast, an immune response against the recipient may cause the devastating graft-versus-host disease (GvHD). Recovery of the immune system (both qualitative and quantitative) after SCT is perhaps the most important factor in determining the clinical outcome. Consequently, immune reconstitution has been extensively studied using different approaches, including quantitative analysis of immune cells as well as their phenotypic characterization. Analysis of diversity and clonality is an important tool in determining competence of the immune system, assuming that a broad diversity assures efficient response to different stimuli and clonal dominance reflects ongoing, potentially relevant immune responses. Detailed analysis of the immune repertoire through the flow cytometric and molecular study of the T cell receptor repertoire has been applied to gain quantitative and qualitative insights about the T cell immune competence and responsiveness. After SCT, a contraction of the T cell pool and a reduction in T cell receptor diversity is clearly associated with clinical immunodeficiency. Reconstitution of the immune system is often characterized by dominance of oligoclonal T cell populations, reflecting specific antigen-driven immune responses. Detailed characterization of T lymphocytes by T cell receptor analysis is possible, and may lead to the identification of individual clones involved in specific immune reactions, such as alloresponses in GvHD, the closely related graft-versus-leukemia effect and opportunistic viral agents such as CMV or EBV.
Collapse
Affiliation(s)
- Christine L O'Keefe
- Experimental Hematology and Hematopoiesis Section, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | | |
Collapse
|
45
|
Pickman Y, Dunn-Walters D, Mehr R. BCR CDR3 length distributions differ between blood and spleen and between old and young patients, and TCR distributions can be used to detect myelodysplastic syndrome. Phys Biol 2013; 10:056001. [PMID: 23965732 DOI: 10.1088/1478-3975/10/5/056001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Complementarity-determining region 3 (CDR3) is the most hyper-variable region in B cell receptor (BCR) and T cell receptor (TCR) genes, and the most critical structure in antigen recognition and thereby in determining the fates of developing and responding lymphocytes. There are millions of different TCR Vβ chain or BCR heavy chain CDR3 sequences in human blood. Even now, when high-throughput sequencing becomes widely used, CDR3 length distributions (also called spectratypes) are still a much quicker and cheaper method of assessing repertoire diversity. However, distribution complexity and the large amount of information per sample (e.g. 32 distributions of the TCRα chain, and 24 of TCRβ) calls for the use of machine learning tools for full exploration. We have examined the ability of supervised machine learning, which uses computational models to find hidden patterns in predefined biological groups, to analyze CDR3 length distributions from various sources, and distinguish between experimental groups. We found that (a) splenic BCR CDR3 length distributions are characterized by low standard deviations and few local maxima, compared to peripheral blood distributions; (b) healthy elderly people's BCR CDR3 length distributions can be distinguished from those of the young; and (c) a machine learning model based on TCR CDR3 distribution features can detect myelodysplastic syndrome with approximately 93% accuracy. Overall, we demonstrate that using supervised machine learning methods can contribute to our understanding of lymphocyte repertoire diversity.
Collapse
Affiliation(s)
- Yishai Pickman
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | | |
Collapse
|
46
|
Tuulasvaara A, Baussand J, Laine P, Paulin L, Salminen J, Auvinen P, Gorochov G, Arstila TP. High-sequence diversity and structural conservation in the human T-cell receptor β junctional region during thymic development. Eur J Immunol 2013; 43:2185-93. [DOI: 10.1002/eji.201343360] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 03/28/2013] [Accepted: 05/08/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Anni Tuulasvaara
- Department of Bacteriology and Immunology; Haartman Institute; University of Helsinki; Helsinki; Finland
| | | | - Pia Laine
- Institute of Biotechnology; University of Helsinki; Helsinki; Finland
| | - Lars Paulin
- Institute of Biotechnology; University of Helsinki; Helsinki; Finland
| | - Jukka Salminen
- Department of Surgery; Hospital for Children and Adolescents; Helsinki University Hospital; Helsinki; Finland
| | - Petri Auvinen
- Institute of Biotechnology; University of Helsinki; Helsinki; Finland
| | | | - T. Petteri Arstila
- Department of Bacteriology and Immunology; Haartman Institute; University of Helsinki; Helsinki; Finland
| |
Collapse
|
47
|
Ringhoffer S, Rojewski M, Döhner H, Bunjes D, Ringhoffer M. T-cell reconstitution after allogeneic stem cell transplantation: assessment by measurement of the sjTREC/βTREC ratio and thymic naive T cells. Haematologica 2013; 98:1600-8. [PMID: 23585532 DOI: 10.3324/haematol.2012.072264] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The immune reconstitution after allogeneic hematopoietic stem cell transplantation comprises thymus-dependent and thymus-independent pathways. We wanted to improve the understanding of this complex process using two different measurements at definite checkpoints of T-cell neogenesis. We therefore assessed the thymus-dependent pathway by combining measurements of single joint T-cell receptor excision circles (sjTREC) and β T-cell receptor excision circles (βTREC) in an improved quantitative light-cycler hybridization polymerase chain reaction assay. In a subgroup of patients, we additionally assessed the proliferation kinetics of the CD31(+) thymic naïve cell population, which corresponds to recent thymic emigrants by six-color immunostaining. After the establishment of normal values in 22 healthy volunteers, we applied our polymerase chain reaction to 66 patients undergoing allogeneic hematopoietic stem cell transplantation at a median age of 44 years. It took more than 2 years after transplant to restore the pre-transplant thymic proliferation capacity. Only one third of the patients in our longitudinal study reached age-adjusted normal values for both sjTREC and βTREC at a median follow-up of 558 days, with acute graft-versus-host disease being the most prominent negative factor by univariate analysis. We observed several patterns of sjTREC and βTREC recovery suggesting different mechanisms of thymic damage in individual patients. In a comparison of CD31(+) thymic naïve cells between volunteers and patients after transplant we found a significantly higher peak proliferation rate within the latter population in the first year after transplantation. The combination of measurements of sjTREC and βTREC by our simplified polymerase chain reaction assay provides insight about the stage of T-cell development affected by different types of damage and may help to choose the correct therapeutic intervention. Besides the sole thymic T-cell neogenesis, proliferation within the CD31(+) thymic naïve cell compartment contributed to the replenishment of the naïve T-cell pool after transplantation.
Collapse
|
48
|
Forcina A, Noviello M, Carbone MR, Bonini C, Bondanza A. Predicting the Clinical Outcome of Allogeneic Hematopoietic Stem Cell Transplantation: The Long and Winding Road toward Validated Immune Biomarkers. Front Immunol 2013; 4:71. [PMID: 23531639 PMCID: PMC3607069 DOI: 10.3389/fimmu.2013.00071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 03/04/2013] [Indexed: 12/04/2022] Open
Abstract
The clinical outcome of allogeneic hematopoietic stem cell transplantation (HSCT) is strongly influenced from the potential complications arising during the delicate phase of post-transplant immune restoration. The quantitative aspects of immune-cell repopulation after HSCT and the qualitative features their functional restitution have been extensively reported. Nevertheless, measurable immune biomarkers predicting the clinical outcome of HSCT await formal validation. The aim of this review is an appraisal of most studies published so far on the predictive value of different T and NK-cell biomarkers after HSCT with emphasis on defined thresholds endorsed by multivariate analysis.
Collapse
Affiliation(s)
- A Forcina
- Experimental Hematology Unit, San Raffaele Scientific Institute Milan, Italy
| | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- Pamela J. Fink
- Department of Immunology, University of Washington, Seattle, Washington 98195;
| |
Collapse
|
50
|
Jain N, Liu H, Artz AS, Anastasi J, Odenike O, Godley LA, Joseph L, Marino S, Kline J, Nguyen V, Schouten V, Kunnavakkam R, Larson RA, Stock W, Ulaszek J, Savage PA, Wickrema A, van Besien K. Immune reconstitution after combined haploidentical and umbilical cord blood transplant. Leuk Lymphoma 2013; 54:1242-9. [PMID: 23088744 DOI: 10.3109/10428194.2012.739688] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Umbilical cord blood (UCB) stem cells are frequently employed for allogeneic stem cell transplant, but delayed myeloid and lymphoid immune reconstitution leads to increased risk of infections. We recently reported the clinical results of 45 patients enrolled on a pilot study combining UCB with a human leukocyte antigen (HLA)-haploidentical donor with reduced-intensity conditioning who showed rapid neutrophil and platelet recovery. We report here preliminary immune reconstitution data of these patients. Patients were assessed for lymphocyte subsets, T-cell diversity, Cylex ImmuKnow assay and serological response to pneumococcal vaccination. Natural killer (NK)-cell and B-cell reconstitution were rapid at 1 month and 3 months, respectively. T-cell recovery was delayed, with a gradual increase in the number of T-cells starting around 6 months post-transplant, and was characterized by a diverse polyclonal T-cell repertoire. Overall, immune reconstitution after haplo-cord transplant is similar to that seen after cord blood transplant, despite infusion of much lower cord blood cell dose.
Collapse
Affiliation(s)
- Nitin Jain
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|