1
|
Maurer K, Antin JH. The graft versus leukemia effect: donor lymphocyte infusions and cellular therapy. Front Immunol 2024; 15:1328858. [PMID: 38558819 PMCID: PMC10978651 DOI: 10.3389/fimmu.2024.1328858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many hematologic malignancies as well as non-malignant conditions. Part of the curative basis underlying HSCT for hematologic malignancies relies upon induction of the graft versus leukemia (GVL) effect in which donor immune cells recognize and eliminate residual malignant cells within the recipient, thereby maintaining remission. GVL is a clinically evident phenomenon; however, specific cell types responsible for inducing this effect and molecular mechanisms involved remain largely undefined. One of the best examples of GVL is observed after donor lymphocyte infusions (DLI), an established therapy for relapsed disease or incipient/anticipated relapse. DLI involves infusion of peripheral blood lymphocytes from the original HSCT donor into the recipient. Sustained remission can be observed in 20-80% of patients treated with DLI depending upon the underlying disease and the intrinsic burden of targeted cells. In this review, we will discuss current knowledge about mechanisms of GVL after DLI, experimental strategies for augmenting GVL by manipulation of DLI (e.g. neoantigen vaccination, specific cell type selection/depletion) and research outlook for improving DLI and cellular immunotherapies for hematologic malignancies through better molecular definition of the GVL effect.
Collapse
Affiliation(s)
| | - Joseph H. Antin
- Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
2
|
Wiercinska E, Quade-Lyssy P, Hümmer C, Beifuß J, Akarkach K, Poppe C, Olevska V, Dzionek J, Lahnor H, Bosio A, Papanikolaou E, Bonig H. Automatic generation of alloreactivity-reduced donor lymphocytes and hematopoietic stem cells from the same mobilized apheresis product. J Transl Med 2023; 21:849. [PMID: 38007485 PMCID: PMC10675913 DOI: 10.1186/s12967-023-04738-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/17/2023] [Indexed: 11/27/2023] Open
Abstract
INTRODUCTION In vitro or in vivo depletion of alloreactive T cells can facilitate haplo-identical hematopoietic stem cell transplantation (HSCT). Very satisfactory transplant outcomes were thus reported for TCRαβ/CD19-depleted hematopoietic stem/progenitor cell (HSPC) grafts. The current semi-automatic manufacturing process on the CliniMACS Plus, although robust, still requires a significant amount of manual labor to be completed. Towards advancing and further facilitating large scale cell processing, a new TCRαβ/CD19 depletion module combined with the previously described CD45RA depletion module (to serve as allo-reactivity attenuated donor lymphocyte infusion) was established on the CliniMACS Prodigy. METHODS We evaluated six apheresis products from G-CSF-mobilized volunteer donors which were split automatically by the Prodigy, one portion each depleted of CD45RA+ or of TCRαβ+ and CD19+ cells. We investigated critical quality attributes for both products. Products were assessed for recovery of HSPCs and mature subsets, as well as depletion efficiency of targeted cells using flow cytometry. Effects of apheresis and product age post 48 h storage at 2-6 °C as well as freeze-thawing on product viability and recovery of WBC and HPSCs were assessed by flow cytometry. RESULTS Ten sequential automatic processes were completed with minimal hands-on time beyond tubing set installation. Depletion efficiency of CD45RA+ resp. TCRαβ+ and CD19+ cells was equivalent to previous reports, achieving mean depletions of 4 log of targeted cells for both products. HSPC products retained TCRγδ+ and NK cells. 48 h storage of apheresis product was associated with the expected modest loss of HSPCs, but depletions remained efficient. Depleted products were stable until at least 72 h after apheresis with stem cell viabilities > 90%. Freeze-thawing resulted in loss of NK cells; post-thaw recovery of viable CD45+ and HSPCs was > 70% and in line with expectation. CONCLUSION The closed, GMP-compatible process generates two separate medicinal products from the same mobilized apheresis product. The CD45RA-depleted products contained functional memory T cells, whereas the TCRαβ/CD19-depleted products included HSPCs, TCRγδ+ and NK cells. Both products are predicted to be effectively depleted of GVH-reactivity while providing immunological surveillance, in support of haplo-identical HSCT.
Collapse
Affiliation(s)
- E Wiercinska
- Department of Cellular Therapeutics (GMP), German Red Cross Blood Service BaWü-He, Institute Frankfurt, Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
| | - P Quade-Lyssy
- Department of Cellular Therapeutics (GMP), German Red Cross Blood Service BaWü-He, Institute Frankfurt, Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
| | - C Hümmer
- Department of Cellular Therapeutics (GMP), German Red Cross Blood Service BaWü-He, Institute Frankfurt, Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
| | - J Beifuß
- Department of Cellular Therapeutics (GMP), German Red Cross Blood Service BaWü-He, Institute Frankfurt, Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
| | - K Akarkach
- Department of Cellular Therapeutics (GMP), German Red Cross Blood Service BaWü-He, Institute Frankfurt, Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
| | - C Poppe
- Department of Cellular Therapeutics (GMP), German Red Cross Blood Service BaWü-He, Institute Frankfurt, Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
| | - V Olevska
- Miltenyi Biotec B.V. & CO. KG, Bergisch Gladbach, Germany
| | - J Dzionek
- Miltenyi Biotec B.V. & CO. KG, Bergisch Gladbach, Germany
| | - H Lahnor
- Miltenyi Biomedicine GmbH, Bergisch Gladbach, Germany
| | - A Bosio
- Miltenyi Biotec B.V. & CO. KG, Bergisch Gladbach, Germany
| | - E Papanikolaou
- Miltenyi Biotec B.V. & CO. KG, Bergisch Gladbach, Germany
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Halvard Bonig
- Department of Cellular Therapeutics (GMP), German Red Cross Blood Service BaWü-He, Institute Frankfurt, Frankfurt, Germany.
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany.
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.
- DRK-BSD BaWüHe, Sandhofstraße 1, 60528, Frankfurt, Germany.
| |
Collapse
|
3
|
Zelikson V, Sabo R, Serrano M, Aqeel Y, Ward S, Al Juhaishi T, Aziz M, Krieger E, Simmons G, Roberts C, Reed J, Buck G, Toor A. Allogeneic haematopoietic cell transplants as dynamical systems: influence of early-term immune milieu on long-term T-cell recovery. Clin Transl Immunology 2023; 12:e1458. [PMID: 37457614 PMCID: PMC10345185 DOI: 10.1002/cti2.1458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/11/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
Objectives Immune recovery following haematopoietic cell transplantation (HCT) functions as a dynamical system. Reducing the duration of intense immune suppression and augmenting antigen presentation has the potential to optimise T-cell reconstitution, potentially influencing long-term outcomes. Methods Based on donor-derived T-cell recovery, 26 patients were adaptively randomised between mycophenolate mofetil (MMF) administered for 30-day post-transplant with filgrastim for cytokine support (MMF30 arm, N = 11), or MMF given for 15 days with sargramostim (MMF15 arm, N = 15). All patients underwent in vivo T-cell depletion with 5.1 mg kg-1 antithymocyte globulin (administered over 3 days, Day -9 through to Day -7) and received reduced intensity 450 cGy total body irradiation (3 fractions on Day -1 and Day 0). Patients underwent HLA-matched related and unrelated donor haematopoietic cell transplantation (HCT). Results Clinical outcomes were equivalent between the two groups. The MMF15 arm demonstrated superior T-cell, as well as T-cell subset recovery and a trend towards superior T-cell receptor (TCR) diversity in the first month with this difference persisting through the first year. T-cell repertoire recovery was more rapid and sustained, as well as more diverse in the MMF15 arm. Conclusion The long-term superior immune recovery in the MMF15 arm, administered GMCSF, is consistent with a disproportionate impact of early interventions in HCT. Modifying the 'immune-milieu' following allogeneic HCT is feasible and may influence long-term T-cell recovery.
Collapse
Affiliation(s)
- Viktoriya Zelikson
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Roy Sabo
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVAUSA
| | - Myrna Serrano
- Department of Microbiology and ImmunologyVirginia Commonwealth UniversityRichmondVAUSA
| | - Younus Aqeel
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Savannah Ward
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Taha Al Juhaishi
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - May Aziz
- Department of PharmacyVirginia Commonwealth UniversityRichmondVAUSA
| | - Elizabeth Krieger
- Department of PediatricsVirginia Commonwealth UniversityRichmondVAUSA
| | - Gary Simmons
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Catherine Roberts
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Jason Reed
- Department of PhysicsVirginia Commonwealth UniversityRichmondVAUSA
| | - Gregory Buck
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVAUSA
| | - Amir Toor
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Lehigh Valley Topper Cancer InstituteAllentownPAUSA
| |
Collapse
|
4
|
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: 3.5] [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
|
5
|
Removal of CD276 + cells from haploidentical memory T-cell grafts significantly lowers the risk of GVHD. Bone Marrow Transplant 2021; 56:2336-2354. [PMID: 33976380 PMCID: PMC8486669 DOI: 10.1038/s41409-021-01307-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 01/08/2023]
Abstract
Detrimental graft-versus-host disease (GVHD) still remains a major cause of death in hematopoietic stem cell transplantation (HSCT). The recently explored depletion of naive cells from mobilized grafts (CD45RA depletion) has shown considerable promise, yet is unable to eliminate the incidence of GVHD. Analysis of CD45RA-depleted haploidentical mixed lymphocytes culture (haplo-MLC) revealed insufficient suppression of alloresponses in the CD4+ compartment and identified CD276 as a marker for alloreactive memory Th1 T cells. Conclusively, depleting CD276+ cells from CD45RA-depleted haplo-MLC significantly attenuated alloreactivity to recipient cells while increasing antiviral reactivity and maintaining anti-third party reactivity in vitro. To evaluate these findings in vivo, bulk, CD45RA-depleted, or CD45RA/CD276-depleted CD4+ T cells from HLA-DR4negative healthy humans were transplanted into NSG-Ab°DR4 mice, a sensitive human allo-GVHD model. Compellingly, CD45RA/CD276-depleted grafts from HLA-DR4negative donors or in vivo depletion of CD276+ cells after transplant of HLA-DR4negative memory CD4 T cells significantly delay the onset of GVHD symptoms and significantly alleviate its severity in NSG-Ab°DR4 mice. The clinical courses correlated with diminished Th1-cytokine secretion and downregulated CXCR6 expression of engrafted peripheral T cells. Collectively, mismatched HLA-mediated GVHD can be controlled by depleting recipient-specific CD276+ alloreacting T cells from the graft, highlighting its application in haplo-HSCT.
Collapse
|
6
|
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: 8] [Impact Index Per Article: 2.7] [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
|
7
|
Ortí G, Palacio-Garcia C, García-Cadenas I, Sánchez-Ortega I, Jimenez MJ, Azqueta C, Villacampa G, Ferrà C, Parody R, Martino R, Bosch F, Querol S, Valcárcel D. Analysis of Cell Subsets in Donor Lymphocyte Infusions from HLA Identical Sibling Donors after Allogeneic Hematopoietic Cell Transplant. Transplant Cell Ther 2020; 27:53.e1-53.e8. [PMID: 32987150 DOI: 10.1016/j.bbmt.2020.09.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Donor lymphocytes infusions (DLIs) are a major therapeutic approach to treat relapse and mixed chimerism after allogeneic hematopoietic cell transplant (alloHCT). The impact of the composition regarding different cell subsets in the development of graft-versus-host disease (GVHD) is not fully understood. We performed a cell subsets analysis of 56 DLIs from fully HLA-compatible identical matched sibling donors (MSDs) in 36 alloHCT patients and studied its association with GVHD. A median of one DLI was infused per patient. Fourteen patients (38%) developed GVHD. The cell composition analysis of the first DLI (DLI1) showed that a high dose of B cells (P = .03) and CD27+ B cells (P < .01) was associated with GVHD. We identified DLI dose cutoff points for several cell populations above which GVHD was more frequent (CD8+ TN >3 × 106 cells/kg, CD27+ B cells >2.6 × 106/kg, CD27+ NK >0.35 × 106 cells/kg, and mononuclear cells >0.83 × 108/kg). Noteworthy, the proportion of CD4+ naive T cells (TN) or unselected TN was not linked with GVHD and a DLI1 containing a higher dose of regulatory T cells was not protective for GVHD. We studied several transplant clinical variables and did not find any association with GVHD. Altogether, this study provides a comprehensive analysis of the cell populations in a DLI from MSDs and identifies potential key cell subsets, which provides insight for the understanding of GVHD after DLI.
Collapse
Affiliation(s)
- Guillermo Ortí
- Departament de Medicina, Universitat Autonoma de Barcelona, Barcelona, Spain; Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain.
| | - Carles Palacio-Garcia
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Irene García-Cadenas
- Hematology Department, Hospital de la Santa Creu i Sant Pau, José Carreras Leukemia Research Institute and IIB Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Isabel Sánchez-Ortega
- Hematology Department, Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain
| | - María José Jimenez
- Hematology Department, Institut Català d'Oncologia, Hospital Universitari Germans Trias i Pujol, Jose Carreras Research Institute, Barcelona, Spain
| | - Carmen Azqueta
- Cellular Therapy Unit, Banc de Sang i Teixits, Barcelona, Spain
| | - Guillermo Villacampa
- Oncology Data Science (ODysSey) Group, Vall d´Hebron Institute of Oncology, Barcelona, Spain
| | - Christelle Ferrà
- Hematology Department, Institut Català d'Oncologia, Hospital Universitari Germans Trias i Pujol, Jose Carreras Research Institute, Barcelona, Spain
| | - Rocio Parody
- Hematology Department, Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain
| | - Rodrigo Martino
- Hematology Department, Hospital de la Santa Creu i Sant Pau, José Carreras Leukemia Research Institute and IIB Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Bosch
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Sergi Querol
- Cellular Therapy Unit, Banc de Sang i Teixits, Barcelona, Spain
| | - David Valcárcel
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
8
|
Laghmouchi A, Hoogstraten C, Falkenburg JHF, Jedema I. Priming of Allo-HLA-DP-Specific Reactivity from the Naïve T Cell Compartment Is Not Exclusively Mediated by Professional Antigen-Presenting Cells. Biol Blood Marrow Transplant 2020; 26:1257-1265. [PMID: 32165326 DOI: 10.1016/j.bbmt.2020.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 10/24/2022]
Abstract
Allogeneic (allo) stem cell transplantation is applied to patients suffering from hematologic malignancies to replace the diseased hematopoietic system with cells derived from a donor stem cell graft. The majority of 10/10-matched unrelated donors are HLA-DP-mismatched, and this may result in varying degrees of the graft-versus-leukemia (GVL) effect with or without the occurrence of graft-versus-host disease (GVHD). Allo-HLA-reactive T cells are commonly present in the donor T cell repertoire, and thus a very profound alloreactive immune response can be provoked in the HLA-DP-mismatched setting. The magnitude and the diversity of the allo-HLA-DP-specific immune response likely dictates the balance between the occurrence of GVL and/or GVHD after transplantation. To understand the nature of the allo-HLA-DP-specific immune response provoked under different stimulatory conditions, immune responses were induced from both the naïve and memory T cell compartments using either HLA-DP-mismatched professional antigen-presenting cells (APCs) (monocyte-derived dendritic cells [allo-DCs]) or HLA-DP-mismatched nonprofessional APCs (skin-derived fibroblasts [allo-fibroblasts]) as stimulator cells. In this study, we observed that allo-HLA-DP-reactive T cells could be provoked from both the naïve and memory compartments by both types of APCs. However, the magnitude of the allo-HLA-DP-specific immune response was greater when stimulation was performed with allo-DCs. Moreover, we found that the frequency of allo-HLA-DP-reactive T cells was greater in the naïve T cell compartment compared with the memory T cell compartment, but we observed a comparable lineage specificity of these allo-HLA-DP-specific reactivities. Overall, the data from this study illustrate that the presence of professional APCs of recipient origin will mostly dictate the magnitude of the allo-HLA-DP-specific immune response derived from both the naïve and memory T cell compartments, but does not exclusively mediate the induction of these immune responses.
Collapse
Affiliation(s)
- Aicha Laghmouchi
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Conny Hoogstraten
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Inge Jedema
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
9
|
ATIR101 administered after T-cell-depleted haploidentical HSCT reduces NRM and improves overall survival in acute leukemia. Leukemia 2020; 34:1907-1923. [PMID: 32047237 PMCID: PMC7326707 DOI: 10.1038/s41375-020-0733-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/09/2020] [Accepted: 01/29/2020] [Indexed: 12/19/2022]
Abstract
Overcoming graft-versus-host disease (GvHD) without increasing relapse and severe infections is a major challenge after allogeneic hematopoietic stem-cell transplantation (HSCT). ATIR101 is a haploidentical, naïve cell-enriched T-cell product, depleted of recipient-alloreactive T cells to minimize the risk of GvHD and provide graft-versus-infection and -leukemia activity. Safety and efficacy of ATIR101 administered after T-cell-depleted haploidentical HSCT (TCD-haplo + ATIR101) without posttransplant immunosuppressors were evaluated in a Phase 2, multicenter study of 23 patients with acute leukemia and compared with an observational cohort undergoing TCD-haplo alone (n = 35), matched unrelated donor (MUD; n = 64), mismatched unrelated donor (MMUD; n = 37), and umbilical cord blood (UCB; n = 22) HSCT. The primary endpoint, 6-month non-relapse mortality (NRM), was 13% with TCD-haplo + ATIR101. One year post HSCT, TCD-haplo + ATIR101 resulted in lower NRM versus TCD-haplo alone (P = 0.008). GvHD-free, relapse-free survival (GRFS) was higher with TCD-haplo + ATIR101 versus MMUD and UCB (both P < 0.03; 1-year rates: 56.5%, 27.0%, and 22.7%, respectively) and was not statistically different from MUD (1 year: 40.6%). ATIR101 grafts with high third-party reactivity were associated with fewer clinically relevant viral infections. Results suggest that haploidentical, selective donor-cell depletion may eliminate requirements for posttransplant immunosuppressors without increasing GvHD risk, with similar GRFS to MUD. Following these results, a randomized Phase 3 trial versus posttransplant cyclophosphamide had been initiated.
Collapse
|
10
|
[Premature immune senescence and chronic kidney disease: Update and perspectives]. Nephrol Ther 2019; 16:9-18. [PMID: 31848067 DOI: 10.1016/j.nephro.2019.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/12/2019] [Indexed: 12/14/2022]
Abstract
Immune senescence is associated with age-related diseases (i.e. infectious disease, cardiovascular diseases and cancers). Chronic kidney disease patients die prematurely when compared with general population, because of a higher occurrence of infections, cardiovascular events and cancer. These diseases are commonly observed in the elderly population and frequently associated with immune senescence. Indeed, chronic kidney disease causes a premature aging of the T lymphocyte compartment, widely related to a decrease in thymic function, a phenomenon that plays a key role in the onset of age-related diseases in chronic kidney disease patients. The degree of immune senescence also influences patients' outcome after renal transplantation, particularly the risk of acute rejection and infections. Partial reversion of pre-transplant immune senescence is observed for some renal transplant patients. In conclusion, to reduce the increasing incidence of morbidity and mortality of chronic kidney disease patients, a better knowledge of uremia-induced immune senescence would help to pave the way to build clinical studies and promote innovative therapeutic approaches. We believe that therapeutic reversion and immune senescence prevention approaches will be part of the management of chronic kidney disease patients in the future.
Collapse
|
11
|
Fernández L, Fernández A, Mirones I, Escudero A, Cardoso L, Vela M, Lanzarot D, de Paz R, Leivas A, Gallardo M, Marcos A, Romero AB, Martínez-López J, Pérez-Martínez A. GMP-Compliant Manufacturing of NKG2D CAR Memory T Cells Using CliniMACS Prodigy. Front Immunol 2019; 10:2361. [PMID: 31649672 PMCID: PMC6795760 DOI: 10.3389/fimmu.2019.02361] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/19/2019] [Indexed: 12/11/2022] Open
Abstract
Natural killer group 2D (NKG2D) is a natural killer (NK) cell-activating receptor that recognizes different stress-induced ligands that are overexpressed in a variety of childhood and adult tumors. NKG2D chimeric antigen receptor (CAR) T cells have shown potent anticancer effects against different cancer types. A second-generation NKG2D CAR was generated by fusing full-length human NKG2D to 4-1BB costimulatory molecule and CD3ζ signaling domain. Patient-derived CAR T cells show limitations including inability to manufacture CAR T cells from the patients' own T cells, disease progression, and death prior to return of engineered cells. The use of allogeneic T cells for CAR therapy could be an attractive alternative, although undesirable graft vs. host reactions may occur. To avoid such adverse effects, we used CD45RA− memory T cells, a T-cell subset with less alloreactivity, as effector cells to express NKG2D CAR. In this study, we developed a protocol to obtain large-scale NKG2D CAR memory T cells for clinical use by using CliniMACS Prodigy, an automated closed system compliant with Good Manufacturing Practice (GMP) guidelines. CD45RA+ fraction was depleted from healthy donors' non-mobilized apheresis using CliniMACS CD45RA Reagent and CliniMACS Plus device. A total of 108 CD45RA− cells were cultured in TexMACS media supplemented with 100 IU/mL IL-2 and activated at day 0 with T Cell TransAct. Then, we used NKG2D-CD8TM-4-1BB-CD3ζ lentiviral vector for cell transduction (MOI = 2). NKG2D CAR T cells expanded between 10 and 13 days. Final cell products were analyzed to comply with the specifications derived from the quality and complementary controls carried out in accordance with the instructions of the Spanish Regulatory Agency of Medicines and Medical Devices (AEMPS) for the manufacture of investigational advanced therapy medicinal products (ATMPs). We performed four validations. The manufacturing protocol here described achieved large numbers of viable NKG2D CAR memory T cells with elevated levels of NKG2D CAR expression and highly cytotoxic against Jurkat and 531MII tumor target cells. CAR T cell final products met release criteria, except for one showing myc overexpression and another with viral copy number higher than five. Manufacturing of clinical-grade NKG2D CAR memory T cells using CliniMACS Prodigy is feasible and reproducible, widening clinical application of CAR T cell therapies.
Collapse
Affiliation(s)
- Lucía Fernández
- Hematological Malignancies H12O, Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Adrián Fernández
- Hematological Malignancies H12O, Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Isabel Mirones
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Adela Escudero
- Pediatric Molecular Hemato-Oncology Department, Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, Madrid, Spain
| | - Leila Cardoso
- Pediatric Molecular Hemato-Oncology Department, Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, Madrid, Spain
| | - María Vela
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Diego Lanzarot
- Applications Department, Miltenyi Biotec S.L., Madrid, Spain
| | - Raquel de Paz
- Hematology Department, Hospital Universitario La Paz, Madrid, Spain
| | - Alejandra Leivas
- Hematological Malignancies H12O, Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Hematology Department, Hospital Universitario12 de Octubre, Madrid, Spain
| | - Miguel Gallardo
- Hematological Malignancies H12O, Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Hematology Department, Hospital Universitario12 de Octubre, Madrid, Spain
| | - Antonio Marcos
- Hematology Department, Hospital Universitario La Paz, Madrid, Spain
| | - Ana Belén Romero
- Hematology Department, Hospital Universitario La Paz, Madrid, Spain
| | - Joaquín Martínez-López
- Hematological Malignancies H12O, Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Hematology Department, Hospital Universitario12 de Octubre, Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain.,Pediatric Hemato-Oncology Department, Hospital Universitario La Paz, Madrid, Spain
| |
Collapse
|
12
|
Bak S, Tischer S, Dragon A, Ravens S, Pape L, Koenecke C, Oelke M, Blasczyk R, Maecker-Kolhoff B, Eiz-Vesper B. Selective Effects of mTOR Inhibitor Sirolimus on Naïve and CMV-Specific T Cells Extending Its Applicable Range Beyond Immunosuppression. Front Immunol 2018; 9:2953. [PMID: 30619313 PMCID: PMC6304429 DOI: 10.3389/fimmu.2018.02953] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022] Open
Abstract
Cytomegalovirus (CMV) infection/reactivation remains among the most important complications of immunosuppression after transplantation. However, recent clinical observations indicate that mammalian target of rapamycin (mTOR) inhibition with sirolimus may improve the outcome of CMV complications. Underlying mechanisms of this observation, particularly the effect of sirolimus on naïve- and CMV-specific cytotoxic CD8+ T-cell (CMV-CTL) functionality is still undiscovered. Here, the influence of sirolimus on naïve and memory CMV-CTLs was determined by CD3/CD28 crosslinking and alloreactivity assays. After stimulating CMV-CTL with HLA-A*02:01-restricted CMVpp65-peptide loaded artificial antigen-presenting cells (aAPCs), we measured the effect of sirolimus on T-cell proliferation, phenotype, and functionality. Sirolimus significantly improved CMV-specific effector memory T-cell function and negatively influenced naïve T cells. This unique mechanism of action was further characterized by increased secretion of interferon-gamma (IFN-γ), granzyme B (GzB) and enhanced target-cell-dependent cytotoxic capacity of activated CMV-CTLs. Next-generation-sequencing (NGS) was applied to monitor T-cell receptor (TCR)-repertoire dynamics and to verify, that the increased functionality was not related to sirolimus-resistant CTL-clones. Instead, modulation of environmental cues during CMV-CTL development via IL-2 receptor (IL-2R)-driven signal transducer and activator of transcription-5 (STAT-5) signaling under mTOR inhibition allowed fine-tuning of T-cell programming for enhanced antiviral response with stable TCR-repertoire dynamics. We show for the first time that sirolimus acts selectively on human naïve and memory T cells and improves CMV-specific T-cell function via modulation of the environmental milieu. The data emphasize the importance to extend immune monitoring including cytokine levels and T-cell functionality which will help to identify patients who may benefit from individually tailored immunosuppression.
Collapse
Affiliation(s)
- Szilvia Bak
- Hannover Medical School, Institute for Transfusion Medicine, Hannover, Germany
| | - Sabine Tischer
- Hannover Medical School, Institute for Transfusion Medicine, Hannover, Germany
| | - Anna Dragon
- Hannover Medical School, Institute for Transfusion Medicine, Hannover, Germany
| | - Sarina Ravens
- Hannover Medical School, Institute of Immunology, Hannover, Germany
| | - Lars Pape
- Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Mathias Oelke
- Department of Pathology, John Hopkins School of Medicine, Baltimore, MD, United States.,NexImmune Inc., Gaithersburg, MD, United States
| | - Rainer Blasczyk
- Hannover Medical School, Institute for Transfusion Medicine, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Hannover Medical School, Institute for Transfusion Medicine, Hannover, Germany
| |
Collapse
|
13
|
Meurer T, Arrieta-Bolaños E, Metzing M, Langer MM, van Balen P, Falkenburg JHF, Beelen DW, Horn PA, Fleischhauer K, Crivello P. Dissecting Genetic Control of HLA-DPB1 Expression and Its Relation to Structural Mismatch Models in Hematopoietic Stem Cell Transplantation. Front Immunol 2018; 9:2236. [PMID: 30344521 PMCID: PMC6183238 DOI: 10.3389/fimmu.2018.02236] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/10/2018] [Indexed: 11/22/2022] Open
Abstract
HLA expression levels have been suggested to be genetically controlled by single nucleotide polymorphisms (SNP) in the untranslated regions (UTR), and expression variants have been associated with the outcome of chronic viral infection and hematopoietic stem cell transplantation (HSCT). In particular, the 3′UTR rs9277534-G/A SNP in HLA-DPB1 has been associated with graft-versus-host-disease after HSCT (Expression model); however its relevance in different immune cells and its mode of action have not been systematically addressed. In addition, there is a strong though not complete overlap between the rs9277534-G/A SNP and structural HLA-DPB1 T cell epitope (TCE) groups which have also been associated with HSCT outcome (TCE Structural model). Here we confirm and extend previous findings of significantly higher HLA-DPB1 expression in B cell lines, unstimulated primary B cells, and monocytes homozygous for rs9277534-G compared to those homozygous for rs9277534-A. However, these differences were abrogated by interferon-γ stimulation or differentiation into dendritic cells. We identify at least seven 3′UTR rs9277534-G/A haplotypes differing by a total of 37 SNP, also characterized by linkage to length variants of a short tandem repeat (STR) in intron 2 and TCE group assignment. 3′UTR mapping did not show any significant differences in post-transcriptional regulation assessed by luciferase assays between two representative rs9277534-G/A haplotypes for any of eight overlapping fragments. Moreover, no evidence for alternative splicing associated with the intron 2 STR was obtained by RT-PCR. In an exemplary cohort of 379 HLA-DPB1 mismatched donor-recipient pairs, risk prediction by the Expression model and the Structural TCE model was 36.7% concordant, with the majority of discordances due to non-applicability of the Expression model. HLA-DPB1 from different TCE groups expressed in the absence of the 3′UTR at similar levels by transfected HeLa cells elicited significantly different mean alloreactive CD4+ T-cell responses, as assessed by CD137 upregulation assays in 178 independent cultures. Taken together, our data provide new insights into the cell type-specific and mechanistic basis of the association between the rs9277534-G/A SNP and HLA-DPB1 expression, and show that, despite partial overlap between both models in HSCT risk-prediction, differential alloreactivity determined by the TCE structural model occurs independently from HLA-DPB1 differential expression.
Collapse
Affiliation(s)
- Thuja Meurer
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany
| | | | - Maximilian Metzing
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany
| | - Mona-May Langer
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Peter van Balen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Dietrich W Beelen
- Department of Bone Marrow Transplantation, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Katharina Fleischhauer
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany.,Deusches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany
| | - Pietro Crivello
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany
| |
Collapse
|
14
|
Approaches to the removal of T-lymphocytes to minimize graft-versus-host disease in patients with primary immunodeficiencies who do not have a matched sibling donor. Curr Opin Allergy Clin Immunol 2018; 17:414-420. [PMID: 28968273 DOI: 10.1097/aci.0000000000000402] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Since the advent of T-lymphocyte depletion in hematopoietic stem cell transplantation (HSCT) for primary immunodeficiency, survival following this procedure has remained poor compared to results when using matched sibling or matched unrelated donors, over the last 40 years. However, three new techniques are radically altering the approach to HSCT for those with no matched donor, particularly those with primary immunodeficiencies which are not severe combined immunodeficiency. RECENT FINDINGS Three main techniques of T-lymphocyte depletion are altering donor choice for patients with primary immunodeficiencies and have improved transplant survival for primary immunodeficiencies to over 90%, equivalent to that for matched sibling and matched unrelated donor transplants. CD3 T cell receptor (TCR)αβ CD19 depletion, CD45RA depletion and use of posttransplant cyclophosphamide give similar overall survival of 90%, although viral reactivation remains a concern. Further modification of CD3 TCRαβ CD19 depletion by adding back inducible caspase-9 suicide gene-modified CD3 TCRαβ T-lymphocytes may further improve outcomes for patients with systemic viral infection. SUMMARY Over the last 5 years, the outcomes of HSCT using new T-lymphocyte depletion methods have improved to the extent that they are equivalent to outcomes of matched sibling donors and may be preferred in the absence of a fully matched sibling donor, over an unrelated donor to reduce the risk of graft versus host disease.
Collapse
|
15
|
Inman CF, Eldershaw SA, Croudace JE, Davies NJ, Sharma-Oates A, Rai T, Pearce H, Sirovica M, Chan YLT, Verma K, Zuo J, Nagra S, Kinsella F, Nunnick J, Amel-Kashipaz R, Craddock C, Malladi R, Moss P. Unique features and clinical importance of acute alloreactive immune responses. JCI Insight 2018; 3:97219. [PMID: 29769441 PMCID: PMC6012511 DOI: 10.1172/jci.insight.97219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 04/19/2018] [Indexed: 01/22/2023] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) can cure some patients with hematopoietic malignancy, but this relies on the development of a donor T cell alloreactive immune response. T cell activity in the first 2 weeks after allo-SCT is crucial in determining outcome, despite the clinical effects of the early alloreactive immune response often not appearing until later. However, the effect of the allogeneic environment on T cells is difficult to study at this time point due to the effects of profound lymphopenia. We approached this problem by comparing T cells at week 2 after allograft to T cells from autograft patients. Allograft T cells were present in small numbers but displayed intense proliferation with spontaneous cytokine production. Oligoclonal expansions at week 2 came to represent a substantial fraction of the established T cell pool and were recruited into tissues affected by graft-versus-host disease. Transcriptional analysis uncovered a range of potential targets for immune manipulation, including OX40L, TWEAK, and CD70. These findings reveal that recognition of alloantigen drives naive T cells toward a unique phenotype. Moreover, they demonstrate that early clonal T cell responses are recruited to sites of subsequent tissue damage and provide a range of targets for potential therapeutic immunomodulation. Alloreactive response T cells at 2 weeks after allo-SCT displayed intense proliferation with spontaneous cytokine production, and were recruited into tissues affected by GvHD.
Collapse
Affiliation(s)
- Charlotte F Inman
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Suzy A Eldershaw
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Joanne E Croudace
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Nathaniel J Davies
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Archana Sharma-Oates
- Centre for Computational Biology, Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Tanuja Rai
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Mirjana Sirovica
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Y L Tracey Chan
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Kriti Verma
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Sandeep Nagra
- Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Francesca Kinsella
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Jane Nunnick
- Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Rasoul Amel-Kashipaz
- Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Charles Craddock
- Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Ram Malladi
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and.,Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and.,Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| |
Collapse
|
16
|
Chabannon C, Kuball J, Bondanza A, Dazzi F, Pedrazzoli P, Toubert A, Ruggeri A, Fleischhauer K, Bonini C. Hematopoietic stem cell transplantation in its 60s: A platform for cellular therapies. Sci Transl Med 2018; 10:10/436/eaap9630. [DOI: 10.1126/scitranslmed.aap9630] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/23/2018] [Indexed: 12/11/2022]
Abstract
Over the last 60 years, more than a million patients received hematopoietic cell transplantation. Having incorporated multiple changes in clinical practices, it remains a complex procedure facing a dual challenge: cure of the underlying disease and prevention of relapse while controlling potentially severe complications. Improved understanding of underlying biological processes resulted in the design of innovative therapies engineered from defined cell populations and testing of these therapies as addition or substitution at virtually every step of the procedure. This review provides an overview of these developments, many of them now applied outside the historical field of hematopoietic cell transplantation.
Collapse
|
17
|
Müller N, Landwehr K, Langeveld K, Stenzel J, Pouwels W, van der Hoorn MA, Seifried E, Bonig H. Generation of alloreactivity-reduced donor lymphocyte products retaining memory function by fully automatic depletion of CD45RA-positive cells. Cytotherapy 2018; 20:532-542. [DOI: 10.1016/j.jcyt.2018.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/19/2017] [Accepted: 01/05/2018] [Indexed: 01/04/2023]
|
18
|
Stikvoort A, Gaballa A, Solders M, Nederlof I, Önfelt B, Sundberg B, Remberger M, Sundin M, Mattsson J, Uhlin M. Risk Factors for Severe Acute Graft-versus-Host Disease in Donor Graft Composition. Biol Blood Marrow Transplant 2018; 24:467-477. [DOI: 10.1016/j.bbmt.2017.11.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/21/2017] [Indexed: 01/17/2023]
|
19
|
Arrieta-Bolaños E, Crivello P, Metzing M, Meurer T, Ahci M, Rytlewski J, Vignali M, Yusko E, van Balen P, Horn PA, Falkenburg JHF, Fleischhauer K. Alloreactive T Cell Receptor Diversity against Structurally Similar or Dissimilar HLA-DP Antigens Assessed by Deep Sequencing. Front Immunol 2018. [PMID: 29520276 PMCID: PMC5827552 DOI: 10.3389/fimmu.2018.00280] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
T cell alloreactivity is mediated by a self-human leukocyte antigen (HLA)-restricted T cell receptor (TCR) repertoire able to recognize both structurally similar and dissimilar allogeneic HLA molecules (i.e., differing by a single or several amino acids in their peptide-binding groove). We hypothesized that thymic selection on self-HLA molecules could have an indirect impact on the size and diversity of the alloreactive response. To test this possibility, we used TCR Vβ immunophenotyping and immunosequencing technology in a model of alloreactivity between self-HLA selected T cells and allogeneic HLA-DPB1 (DPB1) differing from self-DPB1*04:02 by a single (DPB1*02:01) or several (DPB1*09:01) amino acids in the peptide-binding groove. CD4+ T cells from three different self-DPB1*04:01,*04:02 individuals were stimulated with HeLa cells stably transduced with the relevant peptide processing machinery, co-stimulatory molecules, and HLA-DP. Flow cytometric quantification of the DPB1-specific T cell response measured as upregulation of the activation marker CD137 revealed significantly lower levels of alloreactivity against DPB1*02:01 compared with DPB1*09:01 (mean CD4+CD137+ frequency 35.2 ± 9.9 vs. 61.5 ± 7.7%, respectively, p < 0.0001). These quantitative differences were, however, not reflected by differences in the breadth of the alloreactive response at the Vβ level, with both alloantigens eliciting specific responses from all TCR-Vβ specificities tested by flow cytometry, albeit with higher levels of reactivity from most Vβ specificities against DPB1*09:01. In line with these observations, TCRB-CDR3 immunosequencing showed no significant differences in mean clonality of sorted CD137+CD4+ cells alloreactive against DPB1*02:01 or DPB1*09:01 [0.39 (0.36–0.45) and 0.39 (0.30–0.46), respectively], or in the cumulative frequencies of the 10 most frequent responding clones (55–67 and 58–62%, respectively). Most of the clones alloreactive against DPB1*02:01 (68.3%) or DPB1*09:01 (75.3%) were characterized by low-abundance (i.e., they were not appreciable among the pre-culture T cells). Interestingly, however, their cumulative frequency was lower against DPB1*02:01 compared with DPB1*09:01 (mean cumulative frequency 35.3 vs. 50.6%, respectively). Our data show that, despite lower levels of alloreactivity, a similar clonal diversity can be elicited by structurally similar compared with structurally dissimilar HLA-DPB1 alloantigens and demonstrate the power of TCRB immunosequencing in unraveling subtle qualitative changes not appreciable by conventional methods.
Collapse
Affiliation(s)
| | - Pietro Crivello
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany
| | - Maximilian Metzing
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany
| | - Thuja Meurer
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany
| | - Müberra Ahci
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany
| | | | | | - Erik Yusko
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Peter van Balen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | | | - Katharina Fleischhauer
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
20
|
Fernández L, Metais JY, Escudero A, Vela M, Valentín J, Vallcorba I, Leivas A, Torres J, Valeri A, Patiño-García A, Martínez J, Leung W, Pérez-Martínez A. Memory T Cells Expressing an NKG2D-CAR Efficiently Target Osteosarcoma Cells. Clin Cancer Res 2017; 23:5824-5835. [DOI: 10.1158/1078-0432.ccr-17-0075] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/27/2017] [Accepted: 06/21/2017] [Indexed: 11/16/2022]
|
21
|
Cytomegalovirus-Specific T Cells Isolated by IFN-γ Secretion Assay Do Not Induce Significant Graft-Versus-Host Reactions In Vitro. Transplantation 2017; 100:2352-2361. [PMID: 27152919 DOI: 10.1097/tp.0000000000001219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Graft-versus-host (GvH) disease (GvHD) remains a serious concern for patients undergoing antiviral cellular therapy. Despite the major improvements in cellular immunotherapy, the immunogenicity of virus-specific T cells has not yet been fully defined. This present study aims to examine how cytomegalovirus (CMV)-specific cytotoxic T lymphocytes (CTLs) respond to allogeneic antigen stimulation and whether they give rise to GvHD target tissue damage. METHODS Cytomegalovirus-specific CTLs were isolated by the IFN-γ secretion assay (gamma-catch) from healthy seropositive volunteers and expanded in vitro. The levels of intracellular IFN-γ, cytotoxic activity, IFN-γ and granzyme B secretion, and CD25 expression were measured using flow cytometry (fluorescence-activated cell sorting). The ability of CMV-CTLs to induce GvHD target tissue damage was evaluated using the human in vitro skin explant assay (skin explant assay). RESULTS Cytomegalovirus-specific CTLs responded specifically to CMV-phosphoprotein 65 stimulation by secreting IFN-γ and killing virus peptide loaded autologous phytohemagglutinin (PHA) blasts. Compared with unselected peripheral blood mononuclear cells, CMV-CTLs induced significantly less severe cutaneous GvH tissue damage. This observation coincided with low levels of CD25 expression, as well as IFN-γ and granzyme B secretion after allogeneic antigen stimulation in both the mixed lymphocyte reaction and in the skin explant assay. CONCLUSIONS Cytomegalovirus-specific CTLs isolated by the IFN-γ secretion assay from HLA-unmatched healthy donors exhibited a high level of anti-CMV potency without inducing significant cutaneous GvH tissue damage in vitro. This finding provides novel evidence supporting the safe use of in vitro expanded CMV-CTLs as an antiviral therapy in transplant patients with refractory CMV infections.
Collapse
|
22
|
Adoptive immunotherapy for hematological malignancies: Current status and new insights in chimeric antigen receptor T cells. Blood Cells Mol Dis 2016; 62:49-63. [DOI: 10.1016/j.bcmd.2016.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 11/05/2016] [Accepted: 11/06/2016] [Indexed: 12/20/2022]
|
23
|
CD4+ T-cell alloreactivity toward mismatched HLA class II alleles early after double umbilical cord blood transplantation. Blood 2016; 128:2165-2174. [PMID: 27531680 DOI: 10.1182/blood-2016-06-718619] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/09/2016] [Indexed: 11/20/2022] Open
Abstract
Although double umbilical cord blood transplantation (dUCBT) in adult patients may be associated with less graft failure compared with single UCBT, hematopoietic recovery generally originates from a single cord blood unit (CBU). CBU predominance is still incompletely understood. We recently showed that blood CD4+ T-cell numbers rapidly increase after dUCBT, and early CD4+ T-cell chimerism predicts for graft predominance. Given the frequent HLA class II allele mismatches between CBUs in dUCBT, we hypothesized that alloreactive HLA class II-specific CD4+ T cells from the "winning" CBU may contribute to rejection of the "loser" CBU. We evaluated whether CD4+ T cells originating from the predominant (PD)-CBU would recognize HLA class II allele mismatches, expressed by the nonengrafting (NE)-CBU. Alloreactive effector CD4+ T cells toward 1 or more mismatched HLA class II alleles of the NE-CBU were detected in 11 of 11 patients, with reactivity toward 29 of 33 (88%) tested mismatches, and the strongest reactivity toward DR and DQ alleles early after dUCBT. Mismatched HLA class II allele-specific CD4+ T cells recognized primary leukemic cells when the mismatched HLA class II allele was shared between NE-CBU and patient. Our results suggest that cytotoxicity exerted by CD4+ T cells from the PD-CBU drives the rapid rejection of the NE-CBU, whose alloreactive effect might also contribute to graft-versus-leukemia.
Collapse
|
24
|
HLA-DPB1 mismatch alleles represent powerful leukemia rejection antigens in CD4 T-cell immunotherapy after allogeneic stem-cell transplantation. Leukemia 2016; 31:434-445. [DOI: 10.1038/leu.2016.210] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 06/03/2016] [Accepted: 07/04/2016] [Indexed: 12/24/2022]
|
25
|
TNFRSF14 aberrations in follicular lymphoma increase clinically significant allogeneic T-cell responses. Blood 2016; 128:72-81. [PMID: 27103745 DOI: 10.1182/blood-2015-10-679191] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/05/2016] [Indexed: 12/11/2022] Open
Abstract
Donor T-cell immune responses can eradicate lymphomas after allogeneic hematopoietic stem cell transplantation (AHSCT), but can also damage healthy tissues resulting in harmful graft-versus-host disease (GVHD). Next-generation sequencing has recently identified many new genetic lesions in follicular lymphoma (FL). One such gene, tumor necrosis factor receptor superfamily 14 (TNFRSF14), abnormal in 40% of FL patients, encodes the herpes virus entry mediator (HVEM) which limits T-cell activation via ligation of the B- and T-lymphocyte attenuator. As lymphoma B cells can act as antigen-presenting cells, we hypothesized that TNFRSF14 aberrations that reduce HVEM expression could alter the capacity of FL B cells to stimulate allogeneic T-cell responses and impact the outcome of AHSCT. In an in vitro model of alloreactivity, human lymphoma B cells with TNFRSF14 aberrations had reduced HVEM expression and greater alloantigen-presenting capacity than wild-type lymphoma B cells. The increased immune-stimulatory capacity of lymphoma B cells with TNFRSF14 aberrations had clinical relevance, associating with higher incidence of acute GVHD in patients undergoing AHSCT. FL patients with TNFRSF14 aberrations may benefit from more aggressive immunosuppression to reduce harmful GVHD after transplantation. Importantly, this study is the first to demonstrate the impact of an acquired genetic lesion on the capacity of tumor cells to stimulate allogeneic T-cell immune responses which may have wider consequences for adoptive immunotherapy strategies.
Collapse
|
26
|
Bamoulid J, Courivaud C, Crepin T, Carron C, Gaiffe E, Roubiou C, Laheurte C, Moulin B, Frimat L, Rieu P, Mousson C, Durrbach A, Heng AE, Rebibou JM, Saas P, Ducloux D. Pretransplant thymic function predicts acute rejection in antithymocyte globulin-treated renal transplant recipients. Kidney Int 2016; 89:1136-1143. [PMID: 27083287 DOI: 10.1016/j.kint.2015.12.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/26/2015] [Accepted: 12/17/2015] [Indexed: 12/25/2022]
Abstract
Lack of clear identification of patients at high risk of acute rejection hampers the ability to individualize immunosuppressive therapy. Here we studied whether thymic function may predict acute rejection in antithymocyte globulin (ATG)-treated renal transplant recipients in 482 patients prospectively studied during the first year post-transplant of which 86 patients experienced acute rejection. Only CD45RA(+)CD31(+)CD4(+) T cell (recent thymic emigrant [RTE]) frequency (RTE%) was marginally associated with acute rejection in the whole population. This T-cell subset accounts for 26% of CD4(+) T cells. Pretransplant RTE% was significantly associated with acute rejection in ATG-treated patients (hazard ratio, 1.04; 95% confidence interval, 1.01-1.08) for each increased percent in RTE/CD4(+) T cells), but not in anti-CD25 monoclonal (αCD25 mAb)-treated patients. Acute rejection was significantly more frequent in ATG-treated patients with high pretransplant RTE% (31.2% vs. 16.4%) or absolute number of RTE/mm(3) (31.7 vs. 16.1). This difference was not found in αCD25 monclonal antibody-treated patients. Highest values of both RTE% (>31%, hazard ratio, 2.50; 95% confidence interval, 1.09-5.74) and RTE/mm(3) (>200/mm(3), hazard ratio, 3.71; 95% confidence interval, 1.59-8.70) were predictive of acute rejection in ATG-treated patients but not in patients having received αCD25 monoclonal antibody). Results were confirmed in a retrospective cohort using T-cell receptor excision circle levels as a marker of thymic function. Thus, pretransplant thymic function predicts acute rejection in ATG-treated patients.
Collapse
Affiliation(s)
- Jamal Bamoulid
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - Cécile Courivaud
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - Thomas Crepin
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - Clémence Carron
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France
| | - Emilie Gaiffe
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France; CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France
| | - Caroline Roubiou
- Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - Caroline Laheurte
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Bruno Moulin
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Strasbourg, Strasbourg, France
| | - Luc Frimat
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Nancy, Nancy, France
| | - Philippe Rieu
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Reims, Reims, France
| | - Christiane Mousson
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Dijon, Dijon, France
| | - Antoine Durrbach
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Kremlin-Bicêtre, Le Kremlin-Bicêtre, France
| | - Anne-Elisabeth Heng
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Jean-Michel Rebibou
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Dijon, Dijon, France
| | - Philippe Saas
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France; EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Didier Ducloux
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France; CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France.
| |
Collapse
|
27
|
Fleischhauer K, Beelen DW. HLA mismatching as a strategy to reduce relapse after alternative donor transplantation. Semin Hematol 2016; 53:57-64. [PMID: 27000727 DOI: 10.1053/j.seminhematol.2016.01.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Human leukocyte antigen (HLA) mismatches are targets of alloreactive T cells, mediators of graft-versus-leukemia (GvL) and graft-versus-host disease (GvHD) after alternative donor transplantation. Exploitation of HLA mismatching in order to reduce relapse is hampered by necessary interventions aimed at controlling GvHD on the one hand, and by the possibility of immune escape through selective loss of mismatched HLA in relapsing leukemia on the other. Retrospective studies reporting the impact of HLA mismatches on post-transplant relapse need to be interpreted with caution, due to many confounding factors, including disease and use of T-cell depletion, and to be constantly updated to the rapidly changing clinical protocols. Current evidence suggests similar relapse rates for 8/8, 7/8 HLA-matched unrelated, T-cell-replete haploidentical and umbilical cord blood transplantation; however, investigations of locus-specific effects are still scarce in the latter two settings. In unrelated transplantation, a specific role for mismatches at HLA-C and HLA-DPB1, and therein of permissive mismatches defined on the basis of T-cell alloreactivity and/or expression levels, in reducing relapse has been demonstrated in independent studies. This observation suggests new approaches to utilize HLA matching in unrelated donor searches, and the need for further research in the field.
Collapse
Affiliation(s)
- Katharina Fleischhauer
- Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| | - Dietrich W Beelen
- Department for Bone Marrow Transplantation, West-German Cancer Center, University Hospital Essen, Essen, Germany
| |
Collapse
|
28
|
Abstract
Primary immunodeficiencies are rare, inborn errors that result in impaired, disordered or uncontrolled immune responses. Whilst symptomatic and prophylactic treatment is available, hematopoietic stem cell transplantation is an option for many diseases, leading to cure of the immunodeficiency and establishing normal physical and psychological health. Newborn screening for some diseases, whilst improving outcomes, is focusing research on safer and less toxic treatment strategies, which result in durable and sustainable immune function without adverse effects. New conditioning regimens have reduced the risk of hematopoietic stem cell transplantation, and new methods of manipulating stem cell sources should guarantee a donor for almost all patients. Whilst incremental enhancements in transplantation technique have gradually improved survival outcomes over time, some of these new applications are likely to radically alter our approach to treating primary immunodeficiencies.
Collapse
Affiliation(s)
- Andrew Gennery
- Paediatric Immunology and Haematopoietic Stem Cell Transplantation, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK; Paediatric Immunology and Haematopoietic Stem Cell Transplantation, Great North Childrens' Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
29
|
T-cell alloreactivity and transplantation outcome: a budding role for heterologous immunity? Curr Opin Organ Transplant 2015; 20:454-60. [PMID: 26126194 DOI: 10.1097/mot.0000000000000218] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Despite the association between alloreactive T cells and poor graft survival, the mechanisms behind T-cell-mediated rejection are still under investigation. In this review, we will discuss the latest insights into the impact of T-cell alloreactivity on solid organ transplantation and hematopoietic stem cell transplantation (HSCT), with special emphasis on the potential impact of heterologous immunity. RECENT FINDINGS A large part of the memory T-cell repertoire is induced upon virus infections, and evidence for a role of T-cell receptor cross-reactivity of virus-induced memory T cells against allogeneic human leukocyte antigen (HLA) is accumulating in experimental and clinical solid organ transplantation studies. In HSCT, strong alloreactive potential of naïve T cells causes concerns for graft-versus-host disease while additional HLA-DP matching is suggested to prevent CD4 alloreactivity. Furthermore, virus-induced memory T cells hamper mixed chimerism induction, pointing once more towards a role for heterologous immunity. SUMMARY Both memory and naïve T cells contribute to the alloimmune response after transplantation. Monitoring for T-cell phenotypes could help predict rejection episodes and/or graft-versus-host disease, allowing timely intervention. Tailoring donor lymphocyte infusions and additional HLA matching could prevent strong alloreactivity in HSCT. Furthermore, the potential role of heterologous immunity in T-cell alloreactivity and transplantation is gaining interest.
Collapse
|
30
|
Roberto A, Castagna L, Zanon V, Bramanti S, Crocchiolo R, McLaren JE, Gandolfi S, Tentorio P, Sarina B, Timofeeva I, Santoro A, Carlo-Stella C, Bruno B, Carniti C, Corradini P, Gostick E, Ladell K, Price DA, Roederer M, Mavilio D, Lugli E. Role of naive-derived T memory stem cells in T-cell reconstitution following allogeneic transplantation. Blood 2015; 125:2855-64. [PMID: 25742699 PMCID: PMC4424633 DOI: 10.1182/blood-2014-11-608406] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/28/2015] [Indexed: 12/31/2022] Open
Abstract
Early T-cell reconstitution following allogeneic transplantation depends on the persistence and function of T cells that are adoptively transferred with the graft. Posttransplant cyclophosphamide (pt-Cy) effectively prevents alloreactive responses from unmanipulated grafts, but its effect on subsequent immune reconstitution remains undetermined. Here, we show that T memory stem cells (TSCM), which demonstrated superior reconstitution capacity in preclinical models, are the most abundant circulating T-cell population in the early days following haploidentical transplantation combined with pt-Cy and precede the expansion of effector cells. Transferred naive, but not TSCM or conventional memory cells preferentially survive cyclophosphamide, thus suggesting that posttransplant TSCM originate from naive precursors. Moreover, donor naive T cells specific for exogenous and self/tumor antigens persist in the host and contribute to peripheral reconstitution by differentiating into effectors. Similarly, pathogen-specific memory T cells generate detectable recall responses, but only in the presence of the cognate antigen. We thus define the cellular basis of T-cell reconstitution following pt-Cy at the antigen-specific level and propose to explore naive-derived TSCM in the clinical setting to overcome immunodeficiency. These trials were registered at www.clinicaltrials.gov as #NCT02049424 and #NCT02049580.
Collapse
Affiliation(s)
- Alessandra Roberto
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Luca Castagna
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Veronica Zanon
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Stefania Bramanti
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Roberto Crocchiolo
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - James E McLaren
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Sara Gandolfi
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Paolo Tentorio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Barbara Sarina
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Inna Timofeeva
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Armando Santoro
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Carmelo Carlo-Stella
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Benedetto Bruno
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Cristiana Carniti
- Department of Haematology and Pediatric Onco-Haematology, Istituto Nazionale Tumori, Milan, Italy; and
| | - Paolo Corradini
- Department of Haematology and Pediatric Onco-Haematology, Istituto Nazionale Tumori, Milan, Italy; and
| | - Emma Gostick
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Kristin Ladell
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David A Price
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| |
Collapse
|
31
|
High Number of Memory T Cells Is Associated with Higher Risk of Acute Graft-versus-Host Disease after Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2015; 21:569-74. [DOI: 10.1016/j.bbmt.2014.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/11/2014] [Indexed: 11/18/2022]
|
32
|
Chan WK, Suwannasaen D, Throm RE, Li Y, Eldridge PW, Houston J, Gray JT, Pui CH, Leung W. Chimeric antigen receptor-redirected CD45RA-negative T cells have potent antileukemia and pathogen memory response without graft-versus-host activity. Leukemia 2015; 29:387-95. [PMID: 24888271 PMCID: PMC4275423 DOI: 10.1038/leu.2014.174] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/23/2014] [Indexed: 12/27/2022]
Abstract
Chimeric antigen receptor (CAR)-redirected cellular therapy is an attractive modality for cancer treatment. We hypothesized that allogeneic CAR-engineered CD45RA-negative T cells can control cancer and infection without the risk of graft-versus-host disease (GVHD). We used CD19(+) MLL-rearranged leukemia as prototype because it is an aggressive and generally drug-resistant malignancy. CD45RA(-) cells that were transduced with anti-CD19 CAR containing 4-1BB and CD3ζ signaling domains effectively lysed MLL-rearranged leukemia cell lines and primary blasts in vitro. In a disseminated leukemia mouse model, CAR(+)CD45RA(-) cells significantly reduced leukemia burdens and prolonged overall survival without GVHD. CAR(+) cells were sustainable in blood, and all the treated mice remained leukemia-free even after they were re-challenged with leukemia cells. Despite the transduction process, CD45RA(-) cells retained recall activity both in vitro and in vivo against human pathogens commonly found in cancer patients. In comparison with CD45RA(+) cells, CD45RA(-) cells showed less allogeneic activity in mixed leukocyte reactions and in mouse models. Thus, the use of CAR(+)CD45RA(-) cells can separate GVHD from graft-versus-malignancy effect and infection control. These cells should also be useful in nontransplant settings and may be administered as off-the-shelf third-party cells.
Collapse
Affiliation(s)
- W K Chan
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - D Suwannasaen
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - R E Throm
- Vector Laboratory, Department of Experimental Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Y Li
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - P W Eldridge
- Human Application Laboratory, St Jude Children's Research Hospital, Memphis, TN, USA
| | - J Houston
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - J T Gray
- Vector Laboratory, Department of Experimental Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - C-H Pui
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pediatrics, University of Tennessee, Memphis, TN, USA
| | - W Leung
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pediatrics, University of Tennessee, Memphis, TN, USA
| |
Collapse
|
33
|
Touzot F, Neven B, Dal-Cortivo L, Gabrion A, Moshous D, Cros G, Chomton M, Luby JM, Terniaux B, Magalon J, Picard C, Blanche S, Fischer A, Cavazzana M. CD45RA depletion in HLA-mismatched allogeneic hematopoietic stem cell transplantation for primary combined immunodeficiency: A preliminary study. J Allergy Clin Immunol 2014; 135:1303-9.e1-3. [PMID: 25282016 DOI: 10.1016/j.jaci.2014.08.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/21/2014] [Accepted: 08/12/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Combined immunodeficiencies (CIDs) form a heterogeneous group of inherited conditions that affect the development, function, or both of T cells. The treatment of CIDs with allogeneic hematopoietic stem cell transplantation (HSCT) is complicated by a high incidence of life-threatening infections and an increased risk of graft-versus-host disease (GVHD). OBJECTIVE In view of the growing evidence that alloreactivity is mainly derived from human naive T cells, the selective depletion of naive T cells from allografts might constitute a way of reducing alloreactivity while maintaining memory T-cell responsiveness to pathogens. METHODS Five consecutive patients with CIDs and chronic viral infections underwent an allogeneic, HLA-mismatched HSCT. Given the patients' infection status and the potential risk of severe GVHD in the mismatched setting, the CD34(-) fraction of the allograft was depleted of naive T cells by using magnetic CD45RA beads. RESULTS Engraftment occurred in 4 of the 5 patients. No severe GVHD occurred. In the 4 engrafted patients viral infections were cleared within 2 months of the HSCT, and both cellular and humoral immunity were re-established within a year of the HSCT. An early T-cell response against viral pathogens was documented in 2 patients. CONCLUSION The present pilot study shows that clinical-grade depletion of naive T cells from an allograft through the use of magnetic CD45RA beads seems to be a feasible and efficacious option for the treatment of patients with CIDs at high risk of GVHD, infection, or both in an HLA-mismatched setting.
Collapse
Affiliation(s)
- Fabien Touzot
- Département de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; INSERM UMR1163, Paris, France.
| | - Bénédicte Neven
- Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Unité d'Immunologie-Hématologie et Rhumatologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France; INSERM UMR1163, Paris, France
| | - Liliane Dal-Cortivo
- Département de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Aurélie Gabrion
- Département de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Despina Moshous
- Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Unité d'Immunologie-Hématologie et Rhumatologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France; INSERM UMR1163, Paris, France
| | - Guilhem Cros
- Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Unité d'Immunologie-Hématologie et Rhumatologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Maryline Chomton
- Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Unité d'Immunologie-Hématologie et Rhumatologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Jean-Marc Luby
- Département de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Brigitte Terniaux
- Département de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jérémy Magalon
- Département de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Capucine Picard
- Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Centre d'étude des déficits immunitaires (CEDI), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; INSERM UMR1163, Paris, France
| | - Stéphane Blanche
- Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Unité d'Immunologie-Hématologie et Rhumatologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Alain Fischer
- Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Unité d'Immunologie-Hématologie et Rhumatologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France; INSERM UMR1163, Paris, France; College de France, Paris, France
| | - Marina Cavazzana
- Département de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; INSERM UMR1163, Paris, France
| |
Collapse
|