1
|
Lei YY, Chen XR, Jiang S, Guo M, Yu CL, Qiao JH, Cai B, Ai HS, Wang Y, Hu KX. Mechanisms of thymic repair of in vitro-induced precursor T cells as a haplo-identical HSCT regimen. Transplant Cell Ther 2023:S2666-6367(23)01174-0. [PMID: 36944387 DOI: 10.1016/j.jtct.2023.03.015] [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: 12/14/2022] [Revised: 02/06/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) is currently an effective treatment for malignant hematological disease, but the immune deficiency and severe infection triggered by slow immune reconstitution are the main causes of high mortality and transplant failure. One of these outstanding problems is thymus damage, which is associated with graft-versus-host disease (GVHD), and preconditioning including irradiation and chemotherapy. Therefore, rapid repair of damaged thymus and rapid proliferation of thymus-derived donor T cells after transplantation are key to solving the problem. This study is designed to accelerate the recovery of thymus-derived T cells after transplantation. Wild-type mice with normal immunity were used as recipients in a haplo-HSCT mouse model to mimic clinical haplo-HSCT. A modified cell culture system using Notch ligand Delta4 and IL-7 was established that is capable of inducing and amplifying the differentiation and proliferation of hematopoietic stem cells into precursor T (preT) cells in vitro. Haplo-HSCT protocol included the preT and G-CSF mobilized donor splenic mononuclear cells (MNC) co-infusion or MNC alone. Thymic GVHD, thymic repair, and thymus-derived T cell development were compared in two groups by polychromatic immunofluorescence tracking, flow cytometry and detection of T cell receptor Vβ. The thymus homing and T-cell regeneration of allogenic preT cells were observed. The functions of preT cells in accelerating immune reconstitution, restoring thymic architecture, weakening GVH effects, and enhancing immuno-tolerance after transplantation were demonstrated. Further studies revealed that allogeneic preT cells induced by a culture system containing IL7 and Delta4 highly express ccr9 and RANKL. Interestingly, the RANK expression was promoted after preT cells' thymus homing. These results suggested that the RANK/RANKL pathway may play an important role in thymus homing. Our results provide a potential therapeutic option to optimize haplo-HSCT. It further opened up a new field of T cell therapy for artificial induction of allogeneic precursor T cells in vitro to repair the damaged thymus from irradiation and chemotherapy, and to compensate for the recovery of immune function in patients with immune deficiency caused by multiple reasons.
Collapse
Affiliation(s)
- Yang-Yang Lei
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China.
| | - Xin-Rui Chen
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Shan Jiang
- Anhui medical university, anhui province, China
| | - Mei Guo
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Chang-Lin Yu
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Jian-Hui Qiao
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Bo Cai
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Hui-Sheng Ai
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Yi Wang
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China.
| | - Kai-Xun Hu
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China.
| |
Collapse
|
2
|
In Vitro Human Haematopoietic Stem Cell Expansion and Differentiation. Cells 2023; 12:cells12060896. [PMID: 36980237 PMCID: PMC10046976 DOI: 10.3390/cells12060896] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
The haematopoietic system plays an essential role in our health and survival. It is comprised of a range of mature blood and immune cell types, including oxygen-carrying erythrocytes, platelet-producing megakaryocytes and infection-fighting myeloid and lymphoid cells. Self-renewing multipotent haematopoietic stem cells (HSCs) and a range of intermediate haematopoietic progenitor cell types differentiate into these mature cell types to continuously support haematopoietic system homeostasis throughout life. This process of haematopoiesis is tightly regulated in vivo and primarily takes place in the bone marrow. Over the years, a range of in vitro culture systems have been developed, either to expand haematopoietic stem and progenitor cells or to differentiate them into the various haematopoietic lineages, based on the use of recombinant cytokines, co-culture systems and/or small molecules. These approaches provide important tractable models to study human haematopoiesis in vitro. Additionally, haematopoietic cell culture systems are being developed and clinical tested as a source of cell products for transplantation and transfusion medicine. This review discusses the in vitro culture protocols for human HSC expansion and differentiation, and summarises the key factors involved in these biological processes.
Collapse
|
3
|
Bosticardo M, Notarangelo LD. Human thymus in health and disease: Recent advances in diagnosis and biology. Semin Immunol 2023; 66:101732. [PMID: 36863139 PMCID: PMC10134747 DOI: 10.1016/j.smim.2023.101732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/30/2023] [Accepted: 02/14/2023] [Indexed: 03/04/2023]
Abstract
The thymus is the crucial tissue where thymocytes develop from hematopoietic precursors that originate from the bone marrow and differentiate to generate a repertoire of mature T cells able to respond to foreign antigens while remaining tolerant to self-antigens. Until recently, most of the knowledge on thymus biology and its cellular and molecular complexity have been obtained through studies in animal models, because of the difficulty to gain access to thymic tissue in humans and the lack of in vitro models able to faithfully recapitulate the thymic microenvironment. This review focuses on recent advances in the understanding of human thymus biology in health and disease obtained through the use of innovative experimental techniques (eg. single cell RNA sequencing, scRNAseq), diagnostic tools (eg. next generation sequencing), and in vitro models of T-cell differentiation (artificial thymic organoids) and thymus development (eg. thymic epithelial cell differentiation from embryonic stem cells or induced pluripotent stem cells).
Collapse
Affiliation(s)
- Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Four decades of progress. Blood 2022; 140:665-666. [PMID: 35980683 DOI: 10.1182/blood.2022017211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/08/2022] [Indexed: 11/20/2022] Open
|
5
|
Gaudeaux P, Moirangthem RD, Bauquet A, Simons L, Joshi A, Cavazzana M, Nègre O, Soheili S, André I. T-Cell Progenitors As A New Immunotherapy to Bypass Hurdles of Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2022; 13:956919. [PMID: 35874778 PMCID: PMC9300856 DOI: 10.3389/fimmu.2022.956919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the treatment of preference for numerous malignant and non-malignant hemopathies. The outcome of this approach is significantly hampered by not only graft-versus-host disease (GvHD), but also infections and relapses that may occur because of persistent T-cell immunodeficiency following transplantation. Reconstitution of a functional T-cell repertoire can take more than 1 year. Thus, the major challenge in the management of allogeneic HSCT relies on the possibility of shortening the window of immune deficiency through the acceleration of T-cell recovery, with diverse, self-tolerant, and naïve T cells resulting from de novo thymopoiesis from the donor cells. In this context, adoptive transfer of cell populations that can give rise to mature T cells faster than HSCs while maintaining a safety profile compatible with clinical use is of major interest. In this review, we summarize current advances in the characterization of thymus seeding progenitors, and their ex vivo generated counterparts, T-cell progenitors. Transplantation of the latter has been identified as a worthwhile approach to shorten the period of immune deficiency in patients following allogeneic HSCT, and to fulfill the clinical objective of reducing morbimortality due to infections and relapses. We further discuss current opportunities for T-cell progenitor-based therapy manufacturing, including iPSC cell sources and off-the-shelf strategies. These opportunities will be analyzed in the light of results from ongoing clinical studies involving T-cell progenitors.
Collapse
Affiliation(s)
- Pierre Gaudeaux
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
- Smart Immune, Paris, France
| | - Ranjita Devi Moirangthem
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
| | | | - Laura Simons
- Smart Immune, Paris, France
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Akshay Joshi
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
| | - Marina Cavazzana
- Smart Immune, Paris, France
- Department of Biotherapy, Hôpital Universitaire Necker-Enfants Malades, Groupe Hospitalier Paris Centre, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Paris Cité, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
| | | | | | - Isabelle André
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
| |
Collapse
|
6
|
DL4-μbeads induce T cell lineage differentiation from stem cells in a stromal cell-free system. Nat Commun 2021; 12:5023. [PMID: 34408144 PMCID: PMC8373879 DOI: 10.1038/s41467-021-25245-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/26/2021] [Indexed: 11/08/2022] Open
Abstract
T cells are pivotal effectors of the immune system and can be harnessed as therapeutics for regenerative medicine and cancer immunotherapy. An unmet challenge in the field is the development of a clinically relevant system that is readily scalable to generate large numbers of T-lineage cells from hematopoietic stem/progenitor cells (HSPCs). Here, we report a stromal cell-free, microbead-based approach that supports the efficient in vitro development of both human progenitor T (proT) cells and T-lineage cells from CD34+cells sourced from cord blood, GCSF-mobilized peripheral blood, and pluripotent stem cells (PSCs). DL4-μbeads, along with lymphopoietic cytokines, induce an ordered sequence of differentiation from CD34+ cells to CD34+CD7+CD5+ proT cells to CD3+αβ T cells. Single-cell RNA sequencing of human PSC-derived proT cells reveals a transcriptional profile similar to the earliest thymocytes found in the embryonic and fetal thymus. Furthermore, the adoptive transfer of CD34+CD7+ proT cells into immunodeficient mice demonstrates efficient thymic engraftment and functional maturation of peripheral T cells. DL4-μbeads provide a simple and robust platform to both study human T cell development and facilitate the development of engineered T cell therapies from renewable sources. T cells derived from stem cells can be harnessed for regenerative medicine and cancer immunotherapy, but current technologies limit production and translation. Here, the authors present a serum-free, stromal-cell free DLL4-coated microbead method for the scalable production of T-lineage cells from multiple sources of stem cells.
Collapse
|
7
|
Moirangthem RD, Ma K, Lizot S, Cordesse A, Olivré J, de Chappedelaine C, Joshi A, Cieslak A, Tchen J, Cagnard N, Asnafi V, Rausell A, Simons L, Zuber J, Taghon T, Staal FJT, Pflumio F, Six E, Cavazzana M, Lagresle-Peyrou C, Soheili T, André I. A DL-4- and TNFα-based culture system to generate high numbers of nonmodified or genetically modified immunotherapeutic human T-lymphoid progenitors. Cell Mol Immunol 2021; 18:1662-1676. [PMID: 34117371 PMCID: PMC8245454 DOI: 10.1038/s41423-021-00706-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
Several obstacles to the production, expansion and genetic modification of immunotherapeutic T cells in vitro have restricted the widespread use of T-cell immunotherapy. In the context of HSCT, delayed naïve T-cell recovery contributes to poor outcomes. A novel approach to overcome the major limitations of both T-cell immunotherapy and HSCT would be to transplant human T-lymphoid progenitors (HTLPs), allowing reconstitution of a fully functional naïve T-cell pool in the patient thymus. However, it is challenging to produce HTLPs in the high numbers required to meet clinical needs. Here, we found that adding tumor necrosis factor alpha (TNFα) to a DL-4-based culture system led to the generation of a large number of nonmodified or genetically modified HTLPs possessing highly efficient in vitro and in vivo T-cell potential from either CB HSPCs or mPB HSPCs through accelerated T-cell differentiation and enhanced HTLP cell cycling and survival. This study provides a clinically suitable cell culture platform to generate high numbers of clinically potent nonmodified or genetically modified HTLPs for accelerating immune recovery after HSCT and for T-cell-based immunotherapy (including CAR T-cell therapy).
Collapse
Affiliation(s)
- Ranjita Devi Moirangthem
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Kuiying Ma
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Sabrina Lizot
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Anne Cordesse
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Juliette Olivré
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Corinne de Chappedelaine
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Akshay Joshi
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Agata Cieslak
- grid.412134.10000 0004 0593 9113Laboratory of Onco-Hematology, AP-HP, Hôpital Necker-Enfants Malades., Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Institut Necker-Enfants Malades (INEM), INSERM UMR 1151, Paris, France
| | - John Tchen
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Nicolas Cagnard
- grid.508487.60000 0004 7885 7602Plateforme Bio-informatique, Université Paris Descartes, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS 3633, Paris, France
| | - Vahid Asnafi
- grid.412134.10000 0004 0593 9113Laboratory of Onco-Hematology, AP-HP, Hôpital Necker-Enfants Malades., Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Institut Necker-Enfants Malades (INEM), INSERM UMR 1151, Paris, France
| | - Antonio Rausell
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Clinical Bioinformatics, INSERM UMR 1163, Paris, France
| | - Laura Simons
- grid.412134.10000 0004 0593 9113Department of Biotherapy Clinical Investigation Center, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Julien Zuber
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France ,grid.412134.10000 0004 0593 9113Department of Adult Kidney Transplantation, AP-HP, Hôpital Necker, Paris, France
| | - Tom Taghon
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium ,grid.5342.00000 0001 2069 7798Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Frank J. T. Staal
- grid.10419.3d0000000089452978Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Françoise Pflumio
- grid.7429.80000000121866389Team Niche and Cancer in Hematopoiesis, Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Emmanuelle Six
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Marina Cavazzana
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France ,grid.412134.10000 0004 0593 9113Department of Biotherapy Clinical Investigation Center, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Chantal Lagresle-Peyrou
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France ,grid.412134.10000 0004 0593 9113Department of Biotherapy Clinical Investigation Center, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Tayebeh Soheili
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Isabelle André
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| |
Collapse
|
8
|
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: 19] [Impact Index Per Article: 4.8] [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
|
9
|
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: 2.0] [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
|
10
|
André I, Simons L, Ma K, Moirangthem RD, Diana JS, Magrin E, Couzin C, Magnani A, Cavazzana M. Ex vivo generated human T-lymphoid progenitors as a tool to accelerate immune reconstitution after partially HLA compatible hematopoietic stem cell transplantation or after gene therapy. Bone Marrow Transplant 2020; 54:749-755. [PMID: 31431705 DOI: 10.1038/s41409-019-0599-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prolonged T-cell immunodeficiency following HLA- incompatible hematopoietic stem cell transplantation (HSCT) represents a major obstacle hampering the more widespread use of this approach. Strategies to fasten T-cell reconstitution in this setting are highly warranted as opportunistic infections and an increased risk of relapse account for high rates of morbidity and mortality especially during early month following this type of HSCT. We have implemented a feeder free cell system based on the use of the notch ligand DL4 and cytokines allowing for the in vitro differentiation of human T-Lymphoid Progenitor cells (HTLPs) from various sources of CD34+ hematopoietic stem and precursor cells (HSPCs). Co- transplantion of human T-lymphoid progenitors (HTLPs) and non- manipulated HSPCs into immunodeficient mice successfully accelerated the reconstitution of a polyclonal T-cell repertoire. This review summarizes preclinical data on the use of T-cell progenitors for treatment of post- transplantation immunodeficiency and gives insights into the development of GMP based protocols for potential clinical applications including gene therapy approaches. Future clinical trials implementing this protocol will aim at the acceleration of immune reconstitution in different clinical settings such as SCID and leukemia patients undergoing allogeneic transplantation. Apart from pure cell-therapy approaches, the combination of DL-4 culture with gene transduction protocols will open new perspectives in terms of gene therapy applications for primary immunodeficiencies.
Collapse
Affiliation(s)
- Isabelle André
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France. .,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France. .,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France.
| | - Laura Simons
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Kuiying Ma
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Ranjita Devi Moirangthem
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Jean-Sébastien Diana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Elisa Magrin
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Chloé Couzin
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alessandra Magnani
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marina Cavazzana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
11
|
Baboon envelope LVs efficiently transduced human adult, fetal, and progenitor T cells and corrected SCID-X1 T-cell deficiency. Blood Adv 2020; 3:461-475. [PMID: 30755435 DOI: 10.1182/bloodadvances.2018027508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/13/2019] [Indexed: 01/15/2023] Open
Abstract
T cells represent a valuable tool for treating cancers and infectious and inherited diseases; however, they are mainly short-lived in vivo. T-cell therapies would strongly benefit from gene transfer into long-lived persisting naive T cells or T-cell progenitors. Here we demonstrate that baboon envelope glycoprotein pseudotyped lentiviral vectors (BaEV-LVs) far outperformed other LV pseudotypes for transduction of naive adult and fetal interleukin-7-stimulated T cells. Remarkably, BaEV-LVs efficiently transduced thymocytes and T-cell progenitors generated by culture of CD34+ cells on Delta-like ligand 4 (Dll4). Upon NOD/SCIDγC-/- engraftment, high transduction levels (80%-90%) were maintained in all T-cell subpopulations. Moreover, T-cell lineage reconstitution was accelerated in NOD/SCIDγC-/- recipients after T-cell progenitor injection compared with hematopoietic stem cell transplantation. Furthermore, γC-encoding BaEV-LVs very efficiently transduced Dll4-generated T-cell precursors from a patient with X-linked severe combined immunodeficiency (SCID-X1), which fully rescued T-cell development in vitro. These results indicate that BaEV-LVs are valuable tools for the genetic modification of naive T cells, which are important targets for gene therapy. Moreover, they allowed for the generation of gene-corrected T-cell progenitors that rescued SCID-X1 T-cell development in vitro. Ultimately, the coinjection of LV-corrected T-cell progenitors and hematopoietic stem cells might accelerate T-cell reconstitution in immunodeficient patients.
Collapse
|
12
|
Singh J, Zúñiga-Pflücker JC. Producing proT cells to promote immunotherapies. Int Immunol 2019; 30:541-550. [PMID: 30102361 DOI: 10.1093/intimm/dxy051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 08/08/2018] [Indexed: 12/17/2022] Open
Abstract
T lymphocytes are critical mediators of the adaptive immune system and they can be harnessed as therapeutic agents against pathogens and in cancer immunotherapy. T cells can be isolated and expanded from patients and potentially generated in vitro using clinically relevant systems. An ultimate goal for T-cell immunotherapy is to establish a safe, universal effector cell type capable of transcending allogeneic and histocompatibility barriers. To this end, human pluripotent stem cells offer an advantage in generating a boundless supply of T cells that can be readily genetically engineered. Here, we review emerging T-cell therapeutics, including tumor-infiltrating lymphocytes, chimeric antigen receptors and progenitor T cells (proT cells) as well as feeder cell-free in vitro systems for their generation. Furthermore, we explore their potential for adoption in the clinic and highlight the challenges that must be addressed to increase the therapeutic success of a universal immunotherapy.
Collapse
Affiliation(s)
- Jastaranpreet Singh
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | |
Collapse
|
13
|
Simons L, Cavazzana M, André I. Concise Review: Boosting T-Cell Reconstitution Following Allogeneic Transplantation-Current Concepts and Future Perspectives. Stem Cells Transl Med 2019; 8:650-657. [PMID: 30887712 PMCID: PMC6591542 DOI: 10.1002/sctm.18-0248] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/06/2019] [Indexed: 12/14/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the treatment of choice for a large number of malignant and nonmalignant (inherited) diseases of the hematopoietic system. Nevertheless, non‐HLA identical transplantations are complicated by a severe T‐cell immunodeficiency associated with a high rate of infection, relapse and graft‐versus‐host disease. Initial recovery of T‐cell immunity following HSCT relies on peripheral expansion of memory T cells mostly driven by cytokines. The reconstitution of a diverse, self‐tolerant, and naive T‐cell repertoire, however, may take up to 2 years and crucially relies on the interaction of T‐cell progenitors with the host thymic epithelium, which may be altered by GvHD, age or transplant‐related toxicities. In this review, we summarize current concepts to stimulate reconstitution of a peripheral and polyclonal T‐cell compartment following allogeneic transplantation such as graft manipulation (i.e., T‐cell depletion), transfusion of ex vivo manipulated donor T cells or the exogenous administration of cytokines and growth factors to stimulate host‐thymopoiesis with emphasis on approaches which have led to clinical trials. Particular attention will be given to the development of cellular therapies such as the ex vivo generation of T‐cell precursors to fasten generation of a polyclonal and functional host‐derived T‐cell repertoire. Having been tested so far only in preclinical mouse models, clinical studies are now on the way to validate the efficacy of such T‐cell progenitors in enhancing immune reconstitution following HSCT in various clinical settings. stem cells translational medicine2019;00:1–8
Collapse
Affiliation(s)
- Laura Simons
- Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University-Sorbonne Paris Cité, Imagine Institute, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marina Cavazzana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University-Sorbonne Paris Cité, Imagine Institute, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Isabelle André
- Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University-Sorbonne Paris Cité, Imagine Institute, Paris, France
| |
Collapse
|
14
|
Gene therapy targeting haematopoietic stem cells for inherited diseases: progress and challenges. Nat Rev Drug Discov 2019; 18:447-462. [DOI: 10.1038/s41573-019-0020-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
Dynamics of genetically engineered hematopoietic stem and progenitor cells after autologous transplantation in humans. Nat Med 2018; 24:1683-1690. [DOI: 10.1038/s41591-018-0195-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 08/07/2018] [Indexed: 01/04/2023]
|