1
|
Lum SH, Albert MH, Gilbert P, Sirait T, Algeri M, Muratori R, Fournier B, Laberko A, Karakukcu M, Unal E, Ayas M, Yadav SP, Fisgin T, Elfeky R, Fernandes J, Faraci M, Cole T, Schulz A, Meisel R, Zecca M, Ifversen M, Biffi A, Diana JS, Vallée T, Giardino S, Ersoy GZ, Moshous D, Gennery AR, Balashov D, Bonfim C, Locatelli F, Lankester A, Neven B, Slatter M. Outcomes of HLA-mismatched HSCT with TCRαβ/CD19 depletion or post-HSCT cyclophosphamide for inborn errors of immunity. Blood 2024; 144:565-580. [PMID: 38669631 DOI: 10.1182/blood.2024024038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/27/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
ABSTRACT HLA-mismatched transplants with either in vitro depletion of CD3+ T-cell receptor (TCR)αβ/CD19 (TCRαβ) cells or in vivo T-cell depletion using posttransplant cyclophosphamide (PTCY) have been increasingly used for patients with inborn errors of immunity (IEIs). We performed a retrospective multicenter study via the EBMT registry on 306 children with IEIs undergoing their first transplant between 2010 and 2019 from an HLA-mismatched donor using TCRαβ (n = 167) or PTCY (n = 139). The median age for hematopoietic stem cell transplantation (HSCT) was 1.2 years (range, 0.03-19.6 years). The 3-year overall survival (OS) was 78% (95% confidence interval (CI), 71-84) after TCRαβ and 66% (57-74) after PTCY (P = .013). Pre-HSCT morbidity score (hazard ratio [HR], 2.27; 1.07-4.80, P = .032) and non-busulfan/treosulfan conditioning (HR, 3.12; 1.98-4.92, P < .001) were the only independent predictors of unfavorable OS. The 3-year event-free survival (EFS) was 58% (50%-66%) after TCRαβ and 57% (48%-66%) after PTCY (P = .804). The cumulative incidence of severe acute graft-versus-host disease (GvHD) was higher after PTCY (15%, 9%-21%) than TCRαβ (6%, 2%-9%, P = .007), with no difference in chronic GvHD (PTCY, 11%, 6%-17%; TCRαβ, 7%, 3%-11%, P = .173). The 3-year GvHD-free EFS was 53% (44%-61%) after TCRαβ and 41% (32%-50%) after PTCY (P = .080). PTCY had significantly higher rates of veno-occlusive disease (14.4% vs TCRαβ 4.9%, P = .009), acute kidney injury (12.7% vs 4.6%, P = .032), and pulmonary complications (38.2% vs 24.1%, P = .017). Adenoviremia (18.3% vs PTCY 8.0%, P = .015), primary graft failure (10% vs 5%, P = .048), and second HSCT (17.4% vs 7.9%, P = .023) were significantly higher in TCRαβ. In conclusion, this study demonstrates that both approaches are suitable options in patients with IEIs, although they are characterized by different advantages and outcomes.
Collapse
Affiliation(s)
- Su Han Lum
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Michael H Albert
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | - Mattia Algeri
- Department of Paediatric Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Magna Graecia University, Catanzaro, Italy
| | - Rafaella Muratori
- Pediatric Hematology and Transplantation Unit, Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil
| | - Benjamin Fournier
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Alexandra Laberko
- Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Musa Karakukcu
- Erciyes University, KANKA Pediatric Hematology/Oncology and BMT Hospital, Kayseri, Turkey
| | - Elrem Unal
- Hasan KALYONCU University and Medicalpoint Hospital, Gaziantep, Turkey
| | - Mouhab Ayas
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Tunc Fisgin
- Pediatric Hematology/Oncology and BMT Unit, Altinbas University Faculty of Medicine Medical Park Bahcelievler Hospital, Istanbul, Turkey
| | - Reem Elfeky
- Department of Paediatric Immunology, Great Ormand Street Children's Hospital, London, United Kingdom
| | - Juliana Fernandes
- Stem Cell Transplantation Unit, ITACI-Instituto da Criança-Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
- Hematology and Stem Cell Transplantation Unit, Hospital Israelita Albert Einstein, São Paulo, Brazil
- Hematology and Stem Cell Transplantation Unit, Hospital 9 de Julho, São Paulo, Brazil
| | | | - Theresa Cole
- Department of Allergy and Immunology, Royal Children's Hospital, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Marco Zecca
- Paediatric Haematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marianne Ifversen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Alessandra Biffi
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Women and Child Health Department, University of Padua and Padua University Hospital, Padua, Italy
| | - Jean-Sebastien Diana
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Tanja Vallée
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Gizem Zengin Ersoy
- Pediatric Hematology/Oncology and BMT Unit, Altinbas University Faculty of Medicine Medical Park Bahcelievler Hospital, Istanbul, Turkey
| | - Despina Moshous
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Dmitry Balashov
- Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Carmem Bonfim
- Instituto de Pesquisa Pele Pequeno Príncipe/Faculdades Pequeno Príncipe, Pediatric Blood and Marrow Transplantation Service Hospital Pequeno Príncipe, Curitiba, Brazil
| | - Franco Locatelli
- Department of Paediatric Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Arjan Lankester
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Bénédicte Neven
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Mary Slatter
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
2
|
Glushkova S, Shelikhova L, Voronin K, Pershin D, Vedmedskaya V, Muzalevskii Y, Kazachenok A, Kurnikova E, Radygina S, Ilushina M, Khismatullina R, Maschan A, Maschan M. Impact of Natural Killer Cell-Associated Factors on Acute Leukemia Outcomes after Haploidentical Hematopoietic Stem Cell Transplantation with αβ T Cell Depletion in a Pediatric Cohort. Transplant Cell Ther 2024; 30:435.e1-435.e12. [PMID: 38278183 DOI: 10.1016/j.jtct.2024.01.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
The technique of αβ T cell depletion (αβTCD) is a well-established method of hematopoietic stem cell transplantation (HSCT) for children with acute leukemia owing to the low rates of graft-versus-host disease and nonrelapse mortality (NRM). The graft-versus-leukemia effect is generally ascribed to natural killer (NK) cells conserved within the graft. It is not known whether NK-related factors affect the outcome of αβTCD HSCT, however. The aim of this retrospective study was to explore the impact of NK alloreactivity (based on donor-recipient killer immunoglobulin-like receptor [KIR] mismatch), graft NK cell dose, and blood NK cell recovery on day +30 post-HSCT on the incidences of leukemia relapse and NRM. The pediatric acute leukemia cohort comprised 295 patients who underwent their first HSCT from a haploidentical donor in complete remission. During post hoc analysis, the total cohort was divided into subcohorts by diagnosis (acute lymphoblastic leukemia [ALL]/acute myeloid leukemia [AML]), NK alloreactivity prediction (KIR match/KIR mismatch), graft NK cell dose (less than versus greater than the median value), and blood NK cell recovery on day +30 post-HSCT (less than versus greater than the median value). We also investigated the influence of serotherapy (antithymocyte globulin [ATG] group) versus abatacept + tocilizumab combination [aba+toci] group) on relapse risk in the context of KIR mismatch. The risks of relapse and NRM were calculated by the cumulative risk method, and groups were compared using the Gray test. Multivariate analysis revealed no apparent impact of predicted NK alloreactivity or any other studied NK cell-related factors for the entire cohort. For patients with AML, a significantly higher relapse risk associated with high NK cell graft content on the background of no predicted KIR mismatch (P = .002) was shown. Multivariate analysis confirmed this finding (P = .018); on the other hand, for the KIR-mismatched patients, there was a trend toward a lower risk of relapse associated with high NK cell dose. The use of ATG was associated with a trend toward reduced relapse risk (P = .074) in the AML patients. There was no significant impact of NK-related factors in the ALL patients. Overall, the evaluated NK-related factors did not show a clear and straightforward correlation with the key outcomes of HSCT in our cohort of children with acute leukemia. In practice, the data support prioritization of KIR-mismatched donors for patients with AML. Importantly, a potential interaction of KIR ligand mismatch and NK cell content in the graft was identified. Indirect evidence suggests that additional cellular constituents of the graft could influence the function of NK cells after HSCT and affect their role as graft-versus-leukemia effectors.
Collapse
Affiliation(s)
- Svetlana Glushkova
- Laboratory of Transplantation Immunology and Immunotherapy, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| | - Larisa Shelikhova
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Kirill Voronin
- Department of Statistics, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Dmitriy Pershin
- Laboratory of Transplantation Immunology and Immunotherapy, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Viktoria Vedmedskaya
- Laboratory of Transplantation Immunology and Immunotherapy, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Yakov Muzalevskii
- Department of Transfusion Medicine, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexei Kazachenok
- Department of Transfusion Medicine, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Elena Kurnikova
- Department of Transfusion Medicine, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Svetlana Radygina
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Maria Ilushina
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Rimma Khismatullina
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexei Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Michael Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
3
|
Milman Krentsis I, Orgad R, Zheng Y, Bachar Lustig E, Rosen C, Shezen E, Yadav S, Nathansohn Levi B, Assayag M, Berkman N, Karmouty Quintana H, Shoshan E, Blagdon C, Reisner Y. Lung Regeneration by Transplantation of Allogeneic Lung Progenitors Using a Safer Conditioning Regimen and Clinical-grade Reagents. Stem Cells Transl Med 2022; 11:178-188. [PMID: 35298657 PMCID: PMC8929438 DOI: 10.1093/stcltm/szab016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/17/2021] [Indexed: 01/27/2023] Open
Abstract
Over the last decades, several studies demonstrated the possibility of lung regeneration through transplantation of various lung progenitor populations. Recently, we showed in mice that fetal or adult lung progenitors could potentially provide donor cells for transplantation, provided that the lung stem cell niche in the recipient is vacated of endogenous lung progenitors by adequate conditioning. Accordingly, marked lung regeneration could be attained following i.v. infusion of a single cell suspension of lung cells into recipient mice conditioned with naphthalene (NA) and 6Gy total body irradiation (TBI). As clinical translation of this approach requires the use of allogenic donors, we more recently developed a novel transplantation modality based on co-infusion of hematopoietic and lung progenitors from the same donor. Thus, by virtue of hematopoietic chimerism, which leads to immune tolerance toward donor antigens, the lung progenitors can be successfully engrafted without any need for post-transplant immune suppression. In the present study, we demonstrate that it is possible to replace NA in the conditioning regimen with Cyclophosphamide (CY), approved for the treatment of many diseases and that a lower dose of 2 GY TBI can successfully enable engraftment of donor-derived hematopoietic and lung progenitors when CY is administered in 2 doses after the stem cell infusion. Taken together, our results suggest a feasible and relatively safe protocol that could potentially be translated to clinical transplantation of lung progenitors across major MHC barriers in patients with terminal lung diseases.
Collapse
Affiliation(s)
| | | | - Yangxi Zheng
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Esther Bachar Lustig
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Chava Rosen
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel,Department of Neonatology, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tal-Hashomer, Israel
| | - Elias Shezen
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Sandeep Yadav
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Miri Assayag
- Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Neville Berkman
- Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Harry Karmouty Quintana
- University of Texas Health Science Center at Houston, Department of Biochemistry and Molecular Biology & Divisions of Critical Care, Pulmonary and Sleep Medicine, Houston, TX, USA
| | - Einav Shoshan
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Christa Blagdon
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Yair Reisner
- Corresponding author: Yair Reisner, Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Takahashi T, Prockop SE. T-cell depleted haploidentical hematopoietic cell transplantation for pediatric malignancy. Front Pediatr 2022; 10:987220. [PMID: 36313879 PMCID: PMC9614427 DOI: 10.3389/fped.2022.987220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Access to allogenic hematopoietic cell transplantation (HCT), a potentially curative treatment for chemotherapy-resistant hematologic malignancies, can be limited if no human leukocyte antigen (HLA) identical related or unrelated donor is available. Alternative donors include Cord Blood as well as HLA-mismatched unrelated or related donors. If the goal is to minimize the number of HLA disparities, partially matched unrelated donors are more likely to share 8 or 9 of 10 HLA alleles with the recipient. However, over the last decade, there has been success with haploidentical HCT performed using the stem cells from HLA half-matched related donors. As the majority of patients have at least one eligible and motivated haploidentical donor, recruitment of haploidentical related donors is frequently more rapid than of unrelated donors. This advantage in the accessibility has historically been offset by the increased risks of graft rejection, graft-versus-host disease and delayed immune reconstitution. Various ex vivo T-cell depletion (TCD) methods have been investigated to overcome the immunological barrier and facilitate immune reconstitution after a haploidentical HCT. This review summarizes historical and contemporary clinical trials of haploidentical TCD-HCT, mainly in pediatric malignancy, and describes the evolution of these approaches with a focus on serial improvements in the kinetics of immune reconstitution. Methods of TCD discussed include in vivo as well as ex vivo positive and negative selection. In addition, haploidentical TCD as a platform for post-HCT cellular therapies is discussed. The present review highlights that, as a result of the remarkable progress over half a century, haploidentical TCD-HCT can now be considered as a preferred alternative donor option for children with hematological malignancy in need of allogeneic HCT.
Collapse
Affiliation(s)
- Takuto Takahashi
- Pediatric Stem Cell Transplantation, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, United States
| | - Susan E Prockop
- Pediatric Stem Cell Transplantation, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|
5
|
de Witte M, Daenen LGM, van der Wagen L, van Rhenen A, Raymakers R, Westinga K, Kuball J. Allogeneic Stem Cell Transplantation Platforms With Ex Vivo and In Vivo Immune Manipulations: Count and Adjust. Hemasphere 2021; 5:e580. [PMID: 34095763 PMCID: PMC8171366 DOI: 10.1097/hs9.0000000000000580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/14/2021] [Indexed: 01/16/2023] Open
Abstract
Various allogeneic (allo) stem cell transplantation platforms have been developed over the last 2 decades. In this review we focus on the impact of in vivo and ex vivo graft manipulation on immune reconstitution and clinical outcome. Strategies include anti-thymocyte globulin- and post-transplantation cyclophosphamide-based regimens, as well as graft engineering, such as CD34 selection and CD19/αβT cell depletion. Differences in duration of immune suppression, reconstituting immune repertoires, and associated graft-versus-leukemia effects and toxicities mediated through viral reactivations are highlighted. In addition, we discuss the impact of different reconstituting repertoires on donor lymphocyte infusions and post allo pharmacological interventions to enhance tumor control. We advocate for precisely counting all graft ingredients and therapeutic drug monitoring during conditioning in the peripheral blood, and for adjusting dosing accordingly on an individual basis. In addition, we propose novel trial designs to better assess the impact of variations in transplantation platforms in order to better learn from our diversity of "counts" and potential "adjustments." This will, in the future, allow daily clinical practice, strategic choices, and future trial designs to be based on data guided decisions, rather than relying on dogma and habits.
Collapse
Affiliation(s)
- Moniek de Witte
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Laura G. M. Daenen
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Lotte van der Wagen
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Anna van Rhenen
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Reiner Raymakers
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Kasper Westinga
- Cell Therapy Facility, University Medical Center Utrecht, The Netherlands
| | - Jürgen Kuball
- Department of Hematology, University Medical Center Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
6
|
Mukherjee S, Reddy O, Panch S, Stroncek D. Establishment of a cell processing laboratory to support hematopoietic stem cell transplantation and chimeric antigen receptor (CAR)-T cell therapy. Transfus Apher Sci 2021; 60:103066. [PMID: 33472742 DOI: 10.1016/j.transci.2021.103066] [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] [Indexed: 01/17/2023]
Abstract
Cell processing laboratories are an important part of cancer treatment centers. Cell processing laboratories began by supporting hematopoietic stem cell (HSC) transplantation programs. These laboratories adapted closed bag systems, centrifuges, sterile connecting devices and other equipment used in transfusion services/blood banks to remove red blood cells and plasma from marrow and peripheral blood stem cells products. The success of cellular cancer immunotherapies such as Chimeric Antigen Receptor (CAR) T-cells has increased the importance of cell processing laboratories. Since many of the diseases successfully treated by CAR T-cell therapy are also treated by HSC transplantation and since HSC transplantation teams are well suited to manage patients treated with CAR T-cells, many cell processing laboratories have begun to produce CAR T-cells. The methods that have been used to process HSCs have been modified for T-cell enrichment, culture, stimulation, transduction and expansion for CAR T-cell production. While processing laboratories are well suited to manufacture CAR T-cells and other cellular therapies, producing these therapies is challenging. The manufacture of cellular therapies requires specialized facilities which are costly to build and maintain. The supplies and reagents, especially vectors, can also be expensive. Finally, highly skilled staff are required. The use of automated equipment for cell production may reduce labor requirements and the cost of facilities. The steps used to produce CAR T-cells are reviewed, as well as various strategies for establishing a laboratory to manufacture these cells.
Collapse
Affiliation(s)
- Somnath Mukherjee
- Center for Cellular Engineering, Department of Transfusion Medicine, NIH Clinical Center, Bethesda, MD, USA; Department of Transfusion Medicine, All India Institute of Medical Sciences, Bhubaneswar, 751019, Odisha, India
| | - Opal Reddy
- Center for Cellular Engineering, Department of Transfusion Medicine, NIH Clinical Center, Bethesda, MD, USA
| | - Sandhya Panch
- Center for Cellular Engineering, Department of Transfusion Medicine, NIH Clinical Center, Bethesda, MD, USA
| | - David Stroncek
- Center for Cellular Engineering, Department of Transfusion Medicine, NIH Clinical Center, Bethesda, MD, USA.
| |
Collapse
|
7
|
Kwon M, Bailén R, Díez-Martín JL. Evolution of the role of haploidentical stem cell transplantation: past, present, and future. Expert Rev Hematol 2020; 13:835-850. [PMID: 32749913 DOI: 10.1080/17474086.2020.1796621] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The accessibility to haplo-donors has led to an increase in the number of haplo-HSCT worldwide. A systematic search of the PubMed database between 2000 to present was performed. AREAS COVERED In this review, the authors discussed the most used approaches to perform haplo-HSCT and its results: T-cell depletion (TCD, including Perugia platform and its modifications) and T-cell repleted haplo (TCR, including the high-dose post-transplant cyclophosphamide strategy (Baltimore protocol) and the Beijing protocol). The improvements and modifications made to the different strategies have increased the indications of haplo-HSCT, including both malignant and nonmalignant disorders. Focusing on the Baltimore protocol, the authors review the results of the retrospective studies that have compared it to other donor transplants. The limitations of this strategy in terms of toxicity, graft complications, and GVHD are also discussed in detail. Finally, possible approaches to improve the outcomes of TCR haplo-HSCT are presented. EXPERT OPINION The recent advances in the field of haplo-HSCT have allowed a large number of patients with incurable diseases to benefit from this procedure despite not having a matched donor. With all available strategies, virtually no patient who needs an allogeneic transplant should be excluded by the absence of a donor.
Collapse
Affiliation(s)
- Mi Kwon
- Department of Hematology, Hospital General Universitario Gregorio Marañón , Madrid, Spain.,Departement of Translational Oncology, Institute of Health Research Gregorio Marañón , Madrid, Spain
| | - Rebeca Bailén
- Department of Hematology, Hospital General Universitario Gregorio Marañón , Madrid, Spain.,Departement of Translational Oncology, Institute of Health Research Gregorio Marañón , Madrid, Spain
| | - José Luis Díez-Martín
- Department of Hematology, Hospital General Universitario Gregorio Marañón , Madrid, Spain.,Departement of Translational Oncology, Institute of Health Research Gregorio Marañón , Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid , Madrid, Spain
| |
Collapse
|
8
|
Minculescu L, Fischer-Nielsen A, Haastrup E, Ryder LP, Andersen NS, Schjoedt I, Friis LS, Kornblit BT, Petersen SL, Sengelov H, Marquart HV. Improved Relapse-Free Survival in Patients With High Natural Killer Cell Doses in Grafts and During Early Immune Reconstitution After Allogeneic Stem Cell Transplantation. Front Immunol 2020; 11:1068. [PMID: 32547559 PMCID: PMC7273963 DOI: 10.3389/fimmu.2020.01068] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Mature immunocompetent cells from the stem cell graft as well as early robust immune reconstitution are essential for the graft-vs. -tumor (GVT) effect to eliminate residual malignant cells after allogeneic hematopoietic stem cell transplantation (HSCT). In this prospective study we characterized graft composition of T- and NK cell subsets in 88 recipients of peripheral blood stem cell grafts with multicolor flowcytometry. Our primary aim was to analyze the impact of graft composition on immune reconstitution and clinical outcomes after transplantation. Patients transplanted with graft NK cell doses above the median value of 27 × 106/kg had significantly increased relapse-free-survival compared to patients transplanted with lower doses, HR 2.12 (95% CI 1.01-4.45, p = 0.04) Peripheral blood concentrations of NK cells obtained from donors before G-CSF mobilization were significantly correlated to graft NK cell doses (Spearman's ρ 0.53, p = 0.03). The dose of transplanted NK cells/kg correlated significantly with NK cell concentrations in patients early after transplantation (Spearman's ρ 0.26, p = 0.02, and ρ = 0.35, p = 0.001 for days 28 and 56, respectively). Early immune reconstitution above median values of NK cells was significantly associated with improved relapse-free survival (HR 2.84 [95% CI 1.29-6.28], p = 0.01, and HR 4.19 [95% CI 1.68-10.4], p = 0.002, for day 28 and 56, respectively). Early concentrations above the median value of the mature effector CD56dim NK cell subset were significantly associated with decreased relapse incidences at 1 year, 7% (95% CI 1.8-17) vs. 28% (95% CI 15-42), p = 0.04, and 7% (95% CI 1.8-18) vs. 26% (95% CI 14-40) %, p = 0.03, for days 28 and 56, respectively. The results suggest a protective effect of high doses of NK cells in grafts and during early immune reconstitution and support the perception of NK cells as innate effector cells with anti-tumor effects in the setting of allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Lia Minculescu
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Anne Fischer-Nielsen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Eva Haastrup
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Lars Peter Ryder
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Ida Schjoedt
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Lone Smidstrup Friis
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Brian Thomas Kornblit
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Søren Lykke Petersen
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Henrik Sengelov
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Hanne Vibeke Marquart
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
9
|
Kim YE, Kim H, Shin J, Min SY, Kang SH, Suh JK, Koh KN, Im HJ. Stage IV natural killer/T-cell lymphoma with chronic active Epstein-Barr virus, treated with pembrolizumab and TCRαβ-depleted haploidentical hematopoietic stem cell transplantation. Leuk Lymphoma 2020; 61:2250-2253. [PMID: 32352338 DOI: 10.1080/10428194.2020.1757666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yeong Eun Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Republic of Korea
| | - Hyery Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Republic of Korea.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Juhee Shin
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Republic of Korea.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - So Yoon Min
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Republic of Korea.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sung Han Kang
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Republic of Korea.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jin Kyung Suh
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Republic of Korea.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Kyung-Nam Koh
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Republic of Korea.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Ho Joon Im
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Republic of Korea.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
10
|
Gray BP, Requena MD, Nichols MD, Sullenger BA. Aptamers as Reversible Sorting Ligands for Preparation of Cells in Their Native State. Cell Chem Biol 2019; 27:232-244.e7. [PMID: 31879266 DOI: 10.1016/j.chembiol.2019.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/01/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
Although antibodies are routinely used to label and isolate a desired cell type from a more complex mixture of cells, via either fluorescence-activated cell sorting (FACS) or magnetic-activated cell sorting (MACS), such antibody labeling is not easily reversible. We describe an FACS and MACS compatible method to reversibly label and purify cells using aptamers. Magnetic beads loaded with the epidermal growth factor receptor (EGFR)-binding antagonistic aptamer E07 specifically isolated EGFR-expressing cells, and pure, label-free cells were recovered via treatment with an "antidote" oligonucleotide complementary to the aptamer. Additionally, while FACS sorting cells with E07 or EGFR antibody yielded EGFR(+) cells with impeded EGFR signaling, stripping off the aptamer via antidote treatment restored receptor function, returning cells to their native state, which was not possible with the antibody. The ability to reversibly label or isolate cells without compromising their function is a valuable, versatile tool with important implications for both the laboratory and clinic.
Collapse
Affiliation(s)
- Bethany Powell Gray
- Department of Surgery, Duke University Medical Center, 2 Genome Ct, Durham, NC 27710, USA
| | - Martin D Requena
- Department of Surgery, Duke University Medical Center, 2 Genome Ct, Durham, NC 27710, USA
| | - Michael D Nichols
- Department of Biomedical Engineering, Duke University, 101 Science Dr, Durham, NC 27710, USA
| | - Bruce A Sullenger
- Department of Surgery, Duke University Medical Center, 2 Genome Ct, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, 101 Science Dr, Durham, NC 27710, USA.
| |
Collapse
|
11
|
Abstract
Enforced egress of hematopoietic stem cells (HSCs) out of the bone marrow (BM) into the peripheral circulation, termed mobilization, has come a long way since its discovery over four decades ago. Mobilization research continues to be driven by the need to optimize the regimen currently available in the clinic with regard to pharmacokinetic and pharmacodynamic profile, costs, and donor convenience. In this review, we describe the most recent findings in the field and how we anticipate them to affect the development of mobilization strategies in the future. Furthermore, the significance of mobilization beyond HSC collection, i.e. for chemosensitization, conditioning, and gene therapy as well as a means to study the interactions between HSCs and their BM microenvironment, is reviewed. Open questions, controversies, and the potential impact of recent technical progress on mobilization research are also highlighted.
Collapse
Affiliation(s)
- Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, 69120, Germany
| | - Michael P Rettig
- Division of Oncology, Department of Medicine, Washington University School of Medicine,, St. Louis, Missouri, 63110, USA
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine,, St. Louis, Missouri, 63110, USA
| |
Collapse
|
12
|
Aversa F, Pierini A, Ruggeri L, Martelli MF, Velardi A. The Evolution of T Cell Depleted Haploidentical Transplantation. Front Immunol 2019; 10:2769. [PMID: 31827475 PMCID: PMC6890606 DOI: 10.3389/fimmu.2019.02769] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022] Open
Abstract
Work on bone marrow transplantation from haploidentical donor has been proceeding for over 20 years all over the world and new transplant procedures have been developed. To control both graft rejection and graft vs. host disease, some centers have preferred to enhance the intensity of the conditioning regimens and the post-transplant immune suppression in the absence of graft manipulation; others have concentrated on manipulating the graft in the absence of any additional post-transplant immune suppressive agent. Due to the current high engraftment rates, the low incidence of graft-vs.-host disease and regimen related mortality, transplantation from haploidentical donors have been progressively offered even to elderly patients. Overall, survivals compare favorably with reports on transplants from unrelated donors. Further improvements will come with successful implementation of strategies to enhance post-transplant immune reconstitution and to prevent leukemia relapse.
Collapse
Affiliation(s)
- Franco Aversa
- Hematology and Bone Marrow Transplantation Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Antonio Pierini
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Loredana Ruggeri
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Massimo Fabrizio Martelli
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Andrea Velardi
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
13
|
Roldan E, Perales MA, Barba P. Allogeneic Stem Cell Transplantation with CD34+ Cell Selection. Clin Hematol Int 2019; 1:154-160. [PMID: 34595425 PMCID: PMC8432362 DOI: 10.2991/chi.d.190613.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/04/2019] [Indexed: 11/07/2022] Open
Abstract
The success of allogeneic stem cell transplant is hampered by the development of acute and chronic graft-versus-host disease (GvHD) which has direct impact on treatment-related mortality and morbidity. As a result, T cell depletion through positive selection of CD34+ cells has emerged as a promising strategy to reduce acute and chronic GvHD in these patients. In this review, we summarize the main characteristics of allogeneic stem cell transplant with CD34+ cell selection including risks of graft failure, GvHD, infection, organ toxicity, and long-term survival. Moreover, we highlight future strategies to improve the results of this platform and to consolidate its use in clinical practice.
Collapse
Affiliation(s)
- Elisa Roldan
- Hematology Department, Vall d'Hebron University Hospital-Universitat Autónoma de Barcelona, Pg. Vall Hebron 119, Barcelona, Spain
| | - Miguel Angel Perales
- Adult BMT Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pere Barba
- Hematology Department, Vall d'Hebron University Hospital-Universitat Autónoma de Barcelona, Pg. Vall Hebron 119, Barcelona, Spain
| |
Collapse
|
14
|
Bielorai B, Jacoby E, Varda-Bloom N, Hutt D, Churi C, Vernitsky H, Toren A. Haploidentical hematopoietic stem cell transplantation with αβTCR+/CD19+ depletion in pediatric patients with malignant and non-malignant disorders. Bone Marrow Transplant 2019; 54:694-697. [DOI: 10.1038/s41409-019-0607-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
15
|
Haploidentical HSCT for hemoglobinopathies: improved outcomes with TCRαβ +/CD19 +-depleted grafts. Blood Adv 2019; 2:263-270. [PMID: 29431621 DOI: 10.1182/bloodadvances.2017012005] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/28/2017] [Indexed: 11/20/2022] Open
Abstract
We examined outcomes of haploidentical hematopoietic cell transplantation (haplo-HCT) using T-cell receptor αβ+ (TCRαβ+)/CD19+-depleted grafts (TCR group, 14 patients) in children with hemoglobinopathies. Patients received a preparative regimen consisting of busulfan, thiotepa, cyclophosphamide, and antithymocyte globulin preceded by fludarabine, hydroxyurea, and azathioprine. The median follow-up among surviving patients was 3.9 years. The 5-year probabilities of overall survival (OS) and disease-free survival (DFS) were 84% and 69%, respectively. The incidence of graft failure was 14%. We compared outcomes to a historical group of 40 patients with hemoglobinopathies who received CD34+-selected grafts (CD34 group). The median follow-up of surviving patients for the CD34 group was 7.5 years. The 5-year probabilities of OS and DFS were 78% and 39%, respectively. The CD34 group had a significantly higher incidence of graft failure (45%) than the TCR group (14%) (P = .048). The incidences of grades 2 to 4 acute graft-versus-host disease (GVHD) in the TCR and CD34 groups were 28% and 29%, respectively, and 21% and 10% (P = .1), respectively, for extensive chronic GVHD. Viral reactivation was common in both groups. The overall incidence of posttransplant lymphoproliferative disorders for the entire group was 16%. Among all patients, 5 developed autoimmune hemolytic anemia or thrombocytopenia, with the overall cumulative incidence of 11%. The 2 groups showed suboptimal CD4+ recovery within the first 6 months of transplantation with no significant difference between groups. These data demonstrate that TCRαβ+/CD19+-depleted grafts are associated with a reduced incidence of graft failure, but delayed immune reconstitution and associated morbidity and mortality remain a significant challenge.
Collapse
|
16
|
Kim H, Im HJ, Koh KN, Kang SH, Yoo JW, Choi ES, Cho YU, Jang S, Park CJ, Seo JJ. Comparable Outcome with a Faster Engraftment of Optimized Haploidentical Hematopoietic Stem Cell Transplantation Compared with Transplantations from Other Donor Types in Pediatric Acquired Aplastic Anemia. Biol Blood Marrow Transplant 2019; 25:965-974. [PMID: 30639824 DOI: 10.1016/j.bbmt.2019.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/04/2019] [Indexed: 01/31/2023]
Abstract
Haploidentical family donors have been used as an alternative source in hematopoietic cell transplantation for patients with severe aplastic anemia. We evaluated and compared the outcomes of transplantation in pediatric acquired severe aplastic anemia based on donor type. Sixty-seven patients who underwent transplantation between 1998 and 2017 were included. Fourteen patients received grafts from matched sibling donors, 21 from suitable unrelated donors, and 32 from haploidentical family donors. Ex vivo CD3+ or αβ+ T cell-depleted grafts were used for haploidentical transplantation. Sixty-five patients (97.0%) achieved neutrophil engraftment at a median of 11 days. Haploidentical transplantation resulted in significantly faster neutrophil engraftment at a median of 10 days, compared with 14 days in cases of matched sibling donors and 12 days in cases of unrelated donor recipients. Nine patients experienced graft failure, and 5 of 7 who underwent a second transplantation are alive. There was no difference in the incidence of acute or chronic graft-versus-host disease based on donor type. The 5-year overall survival and failure-free survival rates were 93.8% ± 3.0% and 83.3% ± 4.6%, respectively, and there was no significant survival difference based on donor type. The survival outcomes of haploidentical transplantation in patients were comparable with those of matched sibling or unrelated donor transplantation. Optimized haploidentical transplantation using selective T cell depletion and conditioning regimens including low-dose total body irradiation for enhancing engraftment may be a realistic therapeutic option for pediatric patients with severe aplastic anemia.
Collapse
Affiliation(s)
- Hyery Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea
| | - Ho Joon Im
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea.
| | - Kyung-Nam Koh
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea
| | - Sung Han Kang
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea; Department of Pediatrics, Korea University College of Medicine, Seoul, Korea
| | - Jae Won Yoo
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea; Department of Pediatrics, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eun Seok Choi
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea
| | - Young-Uk Cho
- Department of Laboratory Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Seongsoo Jang
- Department of Laboratory Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Chan-Jeoung Park
- Department of Laboratory Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jong Jin Seo
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea
| |
Collapse
|
17
|
Immune Reconstitution Therapy for Immunodeficiency. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
18
|
Abstract
Graft-versus-host (GVHD) is an important cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). As donor T cells are recognized as key drivers of GVHD, some approaches to prevent GVHD have focused on T cell depletion of the allograft. In this review we summarize methods and outcomes of ex vivo T cell depleted (TCD) HCT with a focus on CD34+ selection. This platform is efficacious in preventing acute and chronic GVHD across a wide range of hematologic malignancies, and with the exception of chronic myeloid leukemia, is not associated with adverse relapse or survival outcomes compared to conventional GVHD prophylaxis platforms. In retrospective comparisons recipients of CD34+ selected HCT have higher rates of GVHD-free relapse-free survival (GRFS) than conventional HCT counterparts. Although CD34+ selected allografts require myeloablative and antithymocyte-globulin based conditioning to support engraftment, abrogation of calcineurin inhibitors and methotrexate in this approach reduces its toxicity such that it can be considered in select older and more comorbid patients who could benefit from ablative HCT. A trial comparing GVHD prophylaxis regimens (BMT CTN 1301, NCT02345850) has completed accrual and will be the first to compare CD34+ selected HCT with conventional HCT in a randomized prospective setting. Its findings have potential to establish CD34+ selected HCT as a new standard-of-care platform for GVHD prevention.
Collapse
Affiliation(s)
- Adam R Bryant
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, 10021, USA
| |
Collapse
|
19
|
Haploidentical Bone Marrow Transplantation with Post-Transplantation Cyclophosphamide Plus Thiotepa Improves Donor Engraftment in Patients with Sickle Cell Anemia: Results of an International Learning Collaborative. Biol Blood Marrow Transplant 2018; 25:1197-1209. [PMID: 30500440 DOI: 10.1016/j.bbmt.2018.11.027] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/24/2018] [Indexed: 12/19/2022]
Abstract
Curative therapy for individuals with severe sickle cell disease (SCD) who lack an HLA-identical sibling donor has been frustratingly elusive. In with the goal of improving engraftment while minimizing transplantation-related morbidity, a multi-institutional learning collaborative was developed in the context of a Phase II clinical trial of nonmyeloablative, related HLA-haploidentical (haplo) bone marrow transplantation (BMT) with post-transplantation cyclophosphamide. All eligible participants had hemoglobin SS, and 89% (16 of 18) had an identifiable donor. The median patient age was 20.9 years (IQR, 12.1 to 26.0 years), and the most common indication for transplantation was overt stroke (in 69%; 11 of 16). In the first 3 patients, the conditioning regimen consisted of antithymocyte globulin, fludarabine, cyclophosphamide, and low-dose total body irradiation. Graft-versus-host disease (GVHD) prophylaxis included post-transplantation cyclophosphamide, mycophenolate mofetil, and sirolimus. Primary graft rejection occurred in 2 of the 3 patients (67%), which triggered the study-stopping rule. To reduce graft rejection risk, thiotepa was added to the conditioning regimen, and then 15 patients (including 2 with previous graft rejection) underwent haplo-BMT with this thiotepa-augmented conditioning regimen. At a median follow-up of 13.3 months (interquartile range [IQR], 3.8 to 23.1 months), 93% (14 of 15) had >95% stable donor engraftment at 6 months, with 100% overall survival. The median time to neutrophil engraftment (>500) was 22 days (IQR, 19 to 27 days), and that for platelet engraftment (>50 x 109/L) was 28 days (IQR, 27 days to not reached). Two patients had grade III-IV acute GVHD, 1 patient had mild chronic GVHD, and 86% of patients (6 of 7) were off immunosuppression therapy by 1-year post-transplantation. Our data suggest that haplo-BMT with post-transplantation cyclophosphamide and thiotepa improves donor engraftment without significantly increasing morbidity or mortality and could dramatically expand curative options for individuals with SCD.
Collapse
|
20
|
Salzmann-Manrique E, Bremm M, Huenecke S, Stech M, Orth A, Eyrich M, Schulz A, Esser R, Klingebiel T, Bader P, Herrmann E, Koehl U. Joint Modeling of Immune Reconstitution Post Haploidentical Stem Cell Transplantation in Pediatric Patients With Acute Leukemia Comparing CD34 +-Selected to CD3/CD19-Depleted Grafts in a Retrospective Multicenter Study. Front Immunol 2018; 9:1841. [PMID: 30154788 PMCID: PMC6102342 DOI: 10.3389/fimmu.2018.01841] [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: 03/17/2018] [Accepted: 07/26/2018] [Indexed: 12/25/2022] Open
Abstract
Rapid immune reconstitution (IR) following stem cell transplantation (SCT) is essential for a favorable outcome. The optimization of graft composition should not only enable a sufficient IR but also improve graft vs. leukemia/tumor effects, overcome infectious complications and, finally, improve patient survival. Especially in haploidentical SCT, the optimization of graft composition is controversial. Therefore, we analyzed the influence of graft manipulation on IR in 40 patients with acute leukemia in remission. We examined the cell recovery post haploidentical SCT in patients receiving a CD34+-selected or CD3/CD19-depleted graft, considering the applied conditioning regimen. We used joint model analysis for overall survival (OS) and analyzed the dynamics of age-adjusted leukocytes; lymphocytes; monocytes; CD3+, CD3+CD4+, and CD3+CD8+ T cells; natural killer (NK) cells; and B cells over the course of time after SCT. Lymphocytes, NK cells, and B cells expanded more rapidly after SCT with CD34+-selected grafts (P = 0.036, P = 0.002, and P < 0.001, respectively). Contrarily, CD3+CD4+ helper T cells recovered delayer in the CD34 selected group (P = 0.026). Furthermore, reduced intensity conditioning facilitated faster immune recovery of lymphocytes and T cells and their subsets (P < 0.001). However, the immune recovery for NK cells and B cells was comparable for patients who received reduced-intensity or full preparative regimens. Dynamics of all cell types had a significant influence on OS, which did not differ between patients receiving CD34+-selected and those receiving CD3/CD19-depleted grafts. In conclusion, cell reconstitution dynamics showed complex diversity with regard to the graft manufacturing procedure and conditioning regimen.
Collapse
Affiliation(s)
- Emilia Salzmann-Manrique
- Department of Medicine, Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe-University, Frankfurt, Germany.,Pediatric Hematology and Oncology, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Melanie Bremm
- Pediatric Hematology and Oncology, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Sabine Huenecke
- Pediatric Hematology and Oncology, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Milena Stech
- Pediatric Hematology and Oncology, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Andreas Orth
- University of Applied Sciences Frankfurt, Frankfurt, Germany
| | - Matthias Eyrich
- Pediatric Hematology and Oncology, University of Wuerzburg, Wuerzburg, Germany
| | - Ansgar Schulz
- Pediatric Hematology and Oncology, University of Ulm, Ulm, Germany
| | - Ruth Esser
- Institute of Cellular Therapeutics Hannover Medical School, Hannover, Germany
| | - Thomas Klingebiel
- Pediatric Hematology and Oncology, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Peter Bader
- Pediatric Hematology and Oncology, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Eva Herrmann
- Department of Medicine, Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Ulrike Koehl
- Institute of Cellular Therapeutics Hannover Medical School, Hannover, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany.,Fraunhofer Institute of Cellular Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
21
|
Unexpected High Incidence of Human Herpesvirus-6 Encephalitis after Naive T Cell-Depleted Graft of Haploidentical Stem Cell Transplantation in Pediatric Patients. Biol Blood Marrow Transplant 2018; 24:2316-2323. [PMID: 30031939 DOI: 10.1016/j.bbmt.2018.07.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/10/2018] [Indexed: 11/21/2022]
Abstract
The CD45RA T cell depletion (TCD) method has been used to deplete naive T cells, preventing graft-versus-host disease (GVHD) but preserving memory cells, providing immediate functional T cells with anti-infection, antileukemia, and antirejection effects. We describe a series of 25 consecutive high-risk patients with leukemia who received haploidentical hematopoietic stem cell transplantation (haplo-HSCT) with CD45RA TCD. Each patient received 2 cell products: 1 created by CD34 positive selection and the other through CD45RA depletion from the CD34 negative fraction by a CliniMACS device. CD45RA-depleted haplo-HSCT was well tolerated, with rapid engraftment and low risk of severe acute GVHD and chronic GVHD. Although this treatment achieved a good control of viral reactivations, such as cytomegalovirus and adenovirus, we observed an unexpectedly high rate of limbic encephalitis due to human herpesvirus-6 (HHV-6; 8 cases). Characteristically, the infection appeared early in almost all patients, just after the engraftment. Although no patient died from encephalitis, 1 patient showed neuropsychological sequelae, and another experienced secondary graft failure just after the HHV-6 reactivation.
Collapse
|
22
|
A Conditioning Regimen with Plerixafor Is Safe and Improves the Outcome of TCRαβ+ and CD19+ Cell-Depleted Stem Cell Transplantation in Patients with Wiskott-Aldrich Syndrome. Biol Blood Marrow Transplant 2018; 24:1432-1440. [DOI: 10.1016/j.bbmt.2018.03.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/07/2018] [Indexed: 11/18/2022]
|
23
|
Liu APY, Lee PPW, Kwok JSY, Leung RYY, Chiang AKS, Ha SY, Cheuk DKL, Chan GCF. Selective T cell-depleted haploidentical hematopoietic stem cell transplantation for relapsed/refractory neuroblastoma. Pediatr Transplant 2018; 22:e13240. [PMID: 29921011 DOI: 10.1111/petr.13240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/15/2018] [Indexed: 01/02/2023]
Abstract
Relapsed/refractory NB carries a bleak outcome, warranting novel treatment options. HaploHSCT induces a graft-versus-NB effect via natural killer cell alloreactivity. Review of patients with relapsed/refractory NB who underwent haploHSCT with ex vivo T-cell depletion in our unit from 2013 through 2018. Ten patients were identified (male=5; median age at haploHSCT=6.45 y, range: 3.49-11.02 y). Indications were relapsed in 7 and refractoriness in 3; disease status at haploHSCT was CR in 2, PR in 6, and PD in 2. All patients received peripheral blood stem cell grafts after ex vivo T-cell depletion (CD3/CD19-depletion=1; TCR-αβ/CD19-depletion=4; CD3/CD45RA-depletion=4; and TCR-αβ/CD45RA-depletion=1). Conditioning regimens were fludarabine-based. Neutrophils engrafted on median D + 10 (range: D + 9 to +13), and platelets engrafted (≥20 × 109 /L) on median D + 8 (range: D + 5 to D + 14). Early T- and NK-cell recovery were evident. Of the 10 patients, acute rejection developed in 1 (who died of PD despite rescue HSCT), and 1 died of sepsis before engraftment; 8 experienced full donor-chimerism post-HSCT. Among the 8, 6 experienced CR, 1 died of PD, and 1 died of pulmonary hypertensive crisis before evaluation. At publication, 4 were in remission (2.8, 7.4, 28.5, and 58.9 months). No significant GvHD occurred. HaploHSCT with selective ex vivo T-cell depletion may be a safe and useful salvage strategy for relapsed/refractory NB.
Collapse
Affiliation(s)
- Anthony P Y Liu
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Pamela P W Lee
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Janette S Y Kwok
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong SAR, China
| | - Rock Y Y Leung
- Department of Pathology, Queen Mary Hospital, Hong Kong SAR, China
| | - Alan K S Chiang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Shau-Yin Ha
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Daniel K L Cheuk
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Godfrey C F Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| |
Collapse
|
24
|
Kean LS. Defining success with cellular therapeutics: the current landscape for clinical end point and toxicity analysis. Blood 2018; 131:2630-2639. [PMID: 29728399 PMCID: PMC6032897 DOI: 10.1182/blood-2018-02-785881] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/11/2018] [Indexed: 12/19/2022] Open
Abstract
Cellular therapies play a major and expanding role in the treatment of hematologic diseases. For each of these therapies, a narrow therapeutic window exists, where efficacy is maximized and toxicities minimized. This review focuses on one of the most established cellular therapies, hematopoietic stem cell transplant, and one of the newest cellular therapies, chimeric antigen receptor-T cells. In this review, I will discuss the current state of the field for clinical end point analysis with each of these therapeutics, including their critical toxicities, and focus on the major elements of success for each of these complex treatments for hematologic disease.
Collapse
Affiliation(s)
- Leslie S Kean
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA; Clinical Research Division, The Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Pediatrics, University of Washington, Seattle, WA
| |
Collapse
|
25
|
Erbey F, Akçay A, Atay D, Ovalı E, Öztürk G. Comparison of outcomes after HLA-matched unrelated and αβ T-cell-depleted haploidentical hematopoietic stem cell transplantation for children with high-risk acute leukemia. Pediatr Transplant 2018; 22:e13192. [PMID: 29663666 DOI: 10.1111/petr.13192] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/07/2018] [Indexed: 12/26/2022]
Abstract
T-cell-depleted HAPLO HSCT is an option to treat children with high-risk acute leukemia lacking an HLA-identical donor. We reviewed the outcome of children with acute leukemia after HAPLO (n = 21) and HLA-MUD (n = 32) transplantation. The proportion of patients with ≥CR2 was significantly higher in HAPLO transplantation than MUD transplantation. Patients with MUD transplantation were significantly higher ABO incompatible than patients with HAPLO transplantation. There was no difference between the 2 groups in terms of engraftment, aGvHD and cGvHD, VOD, hemorrhagic cystitis, infections, and relapse. The 5-year OS of MUD transplantation and HAPLO transplantation groups was found 65.8% and 71.1%, respectively (log-rank 0.51). The 5-year RFS was 80.7% for MUD transplantation group and 86.9% for HAPLO transplantation group (log-rank 0.48). There was no statistically significant difference between 2 groups according to TRM (25% MUD transplantation vs 16.3% HAPLO transplantation, log-rank 0.48). These data suggest that survival for patients with high-risk acute leukemia after HAPLO transplantation with ex vivo ɑβ+ T-cell depletion is comparable with MUD transplantation.
Collapse
Affiliation(s)
- Fatih Erbey
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University School of Medicine, Atakent Hospital, İstanbul, Turkey
| | - Arzu Akçay
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University School of Medicine, Atakent Hospital, İstanbul, Turkey
| | - Didem Atay
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University School of Medicine, Atakent Hospital, İstanbul, Turkey
| | - Ercüment Ovalı
- Department of Hematology, Acıbadem University School of Medicine, Altunizade Hospital, İstanbul, Turkey
| | - Gülyüz Öztürk
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University School of Medicine, Atakent Hospital, İstanbul, Turkey
| |
Collapse
|
26
|
Schiller CB, Braciak TA, Fenn NC, Seidel UJE, Roskopf CC, Wildenhain S, Honegger A, Schubert IA, Schele A, Lämmermann K, Fey GH, Jacob U, Lang P, Hopfner KP, Oduncu FS. CD19-specific triplebody SPM-1 engages NK and γδ T cells for rapid and efficient lysis of malignant B-lymphoid cells. Oncotarget 2018; 7:83392-83408. [PMID: 27825135 PMCID: PMC5347777 DOI: 10.18632/oncotarget.13110] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/03/2016] [Indexed: 12/19/2022] Open
Abstract
Triplebodies are antibody-derived recombinant proteins carrying 3 antigen-binding domains in a single polypeptide chain. Triplebody SPM-1 was designed for lysis of CD19-bearing malignant B-lymphoid cells through the engagement of CD16-expressing cytolytic effectors, including NK and γδ T cells. SPM-1 is an optimized version of triplebody ds(19-16-19) and includes humanization, disulfide stabilization and the removal of potentially immunogenic sequences. A three-step chromatographic procedure yielded 1.7 - 5.5 mg of purified, monomeric protein per liter of culture medium. In cytolysis assays with NK cell effectors, SPM-1 mediated potent lysis of cancer-derived B cell lines and primary cells from patients with various B-lymphoid malignancies, which surpassed the ADCC activity of the therapeutic antibody Rituximab. EC50-values ranged from 3 to 86 pM. Finally, in an impedance-based assay, SPM-1 mediated a particularly rapid lysis of CD19-bearing target cells by engaging and activating both primary and expanded human γδ T cells from healthy donors as effectors. These data establish SPM-1 as a useful tool for a kinetic analysis of the cytolytic reactions mediated by γδ T and NK cells and as an agent deserving further development towards clinical use for the treatment of B-lymphoid malignancies.
Collapse
Affiliation(s)
- Christian B Schiller
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-University, Munich, Germany
| | - Todd A Braciak
- Division of Hematology and Oncology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Nadja C Fenn
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-University, Munich, Germany
| | - Ursula J E Seidel
- Department of General Paediatrics, Oncology/Haematology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Claudia C Roskopf
- Division of Hematology and Oncology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Sarah Wildenhain
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-University, Munich, Germany
| | | | - Ingo A Schubert
- Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexandra Schele
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-University, Munich, Germany
| | - Kerstin Lämmermann
- Division of Hematology and Oncology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | | | | | - Peter Lang
- Department of General Paediatrics, Oncology/Haematology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Karl-Peter Hopfner
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-University, Munich, Germany
| | - Fuat S Oduncu
- Division of Hematology and Oncology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
27
|
Jacoby E, Varda-Bloom N, Goldstein G, Hutt D, Churi C, Vernitsky H, Toren A, Bielorai B. Comparison of two cytoreductive regimens for αβ-T-cell-depleted haploidentical HSCT in pediatric malignancies: Improved engraftment and outcome with TBI-based regimen. Pediatr Blood Cancer 2018; 65. [PMID: 28988422 DOI: 10.1002/pbc.26839] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Graft manipulation using selective depletion of αβ-T cells provides a source of haploidentical hematopoietic stem cell transplantation (haplo-HSCT) enriched in effector cells. We report our experience implementing this haplo-HSCT for high-risk malignancies in pediatric patients focusing on the conditioning regimen. PROCEDURE We performed a retrospective study of patients who underwent T-cell receptor αβ-depleted haplo-HSCT for high-risk pediatric malignancies. RESULTS Eighteen patients underwent haplo-HSCT using this method. The initial reduced-toxicity chemotherapy-based conditioning regimen was given to eight patients, and resulted in a high rate of graft rejections (six of eight patients). Thus, total body irradiation (TBI) based regimen was introduced in the following 10 patients and resulted in engraftment in all patients. Neutrophil and platelet engraftment were rapid (median time to engraft, 10 days and 12 days, respectively). Significant treatment-related complications for both cohorts were all due to graft failure in patients receiving chemotherapy-based conditioning, with a treatment-related mortality rate of 17%. None of the patients developed hepatic sinusoidal-obstruction syndrome, and no grade III-IV acute graft versus host disease (GVHD) was observed. The majority of patients were free of immunosuppression in the first 100 days post-HSCT, and only two patients developed chronic GVHD. The cumulative incidence of relapse was 39%. Compared to patients conditioned with chemotherapy, patients conditioned with TBI had superior actuarial overall survival (66% vs. 37%, P = 0.05) and event-free survival (61% vs. 33%, P = 0.04). CONCLUSIONS A TBI-based conditioning for haplo-HSCT using αβ-T-cell depletion for malignant diseases ensured engraftment and resulted in acceptable outcomes.
Collapse
Affiliation(s)
- Elad Jacoby
- Department of Pediatric Hematology, Oncology & BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel.,Institute for Pediatric Research, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Gal Goldstein
- Department of Pediatric Hematology, Oncology & BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel.,Institute for Pediatric Research, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daphna Hutt
- Department of Pediatric Hematology, Oncology & BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Chaim Churi
- Department of Pediatric Hematology, Oncology & BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Helly Vernitsky
- Hematology Laboratory, Sheba Medical Center, Ramat-Gan, Israel
| | - Amos Toren
- Department of Pediatric Hematology, Oncology & BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel.,Institute for Pediatric Research, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Bella Bielorai
- Department of Pediatric Hematology, Oncology & BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel.,Institute for Pediatric Research, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Maschan M, Blagov S, Shelikhova L, Shekhovtsova Z, Balashov D, Starichkova J, Kurnikova E, Boyakova E, Muzalevskii Y, Kazachenok A, Trakhtman P, Osipova E, Khripkova N, Zhogov V, Novichkova G, Maschan A. Low-dose donor memory T-cell infusion after TCR alpha/beta depleted unrelated and haploidentical transplantation: results of a pilot trial. Bone Marrow Transplant 2017; 53:264-273. [PMID: 29269793 DOI: 10.1038/s41409-017-0035-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/18/2017] [Accepted: 09/27/2017] [Indexed: 01/14/2023]
Abstract
Recovery of immunity is delayed in recipients of T-depleted grafts. Adoptive transfer of memory T-cells may improve immune response to common pathogens. A cohort of 53 patients with malignant (n = 36) and non-malignant conditions (n = 17) received TCR alpha/beta depleted grafts from haploidentical (n = 25) or MUD (n = 28) donors. Donor lymphocytes were depleted of CD45RA-positive cells. At a median of 48 days after transplantation, patients received DLI at 25 × 103/kg CD3 cells from haploidentical or 100 × 103/kg CD3 from MUD donors. Up to 3 doses of donor lymphocytes were administered at monthly intervals, escalating to 100 × 103/kg in haploidentical transplants and 300 × 103/kg in MUD transplants. At a median follow-up of 23 months, the cumulative incidence of de novo acute GVHD after DLI is 2% (1 of 43), while the rate of reactivation of preexisting aGVHD was 50% (5 of 10). The transplant-related mortality is 6%. The overall survival rates are 80% and 88% in malignant and non-malignant conditions, respectively. Among patients with absent CMV-specific immune reactivity at baseline (n = 31) expansion of CMV-specific T-cells was demonstrated in 20 (64.5%) within 100 days. Infusions of low dose donor memory T-lymphocytes are safe and constitute a simple measure to prevent infections in the setting of alpha/beta T cell-depleted transplantation.
Collapse
Affiliation(s)
- Michael Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| | - Sergey Blagov
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Larisa Shelikhova
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Zhanna Shekhovtsova
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Dmitriy Balashov
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Julia Starichkova
- Statistics, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Elena Kurnikova
- Transfusion Medicine Service, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Elena Boyakova
- Transplantation Immunology and Immunotherapy Laboratory, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Yakov Muzalevskii
- Transfusion Medicine Service, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexei Kazachenok
- Transfusion Medicine Service, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Pavel Trakhtman
- Transfusion Medicine Service, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Elena Osipova
- Stem Cell Physiology Laboratory, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Natalia Khripkova
- Stem Cell Physiology Laboratory, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Vladimir Zhogov
- Stem Cell Physiology Laboratory, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Galina Novichkova
- Medical Department, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexei Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
29
|
de Witte MA, Kuball J, Miller JS. NK Cells and γδT Cells for Relapse Protection After Allogeneic Hematopoietic Cell Transplantation (HCT). CURRENT STEM CELL REPORTS 2017; 3:301-311. [PMID: 29399441 DOI: 10.1007/s40778-017-0106-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Purpose of review The outcome of allogeneic stem cell transplantation (allo-HCT) is still compromised by relapse and complications. NK cells and γδT cells, effectors which both function through MHC-unrestricted mechanisms, can target transformed and infected cells without inducing Graft-versus-Host Disease (GVHD). Allo-HCT platforms based on CD34+ selection or αβ-TCR depletion result in low grades of GVHD, early immune reconstitution (IR) of NK and γδT cells and minimal usage of GVHD prophylaxis. In this review we will discuss strategies to retain and expand the quantity, diversity and functionality of these reconstituting innate cell types. Recent findings Bisphosphonates, IL-15 cytokine administration, specific antibodies, checkpoint inhibitors and (CMV based) vaccination are currently being evaluated to enhance IR. All these approaches have shown to potentially enhance both NK and γδT cell immuno-repertoires. Summary Rapidly accumulating data linking innate biology to proposed clinical immune interventions, will give unique opportunities to unravel shared pathways which determine the Graft-versus-Tumor effects of NK and γδT cells.
Collapse
Affiliation(s)
- Moniek A de Witte
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN.,Department of Hematology, Cancer Center, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jürgen Kuball
- Department of Hematology, Cancer Center, University Medical Centre Utrecht, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jeffrey S Miller
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN
| |
Collapse
|
30
|
Simonetta F, Masouridi-Levrat S, Beauverd Y, Tsopra O, Tirefort Y, Koutsi A, Stephan C, Polchlopek-Blasiak K, Pradier A, Dantin C, Ansari M, Roosnek E, Chalandon Y. Partial T-cell depletion improves the composite endpoint graft-versus-host disease-free, relapse-free survival after allogeneic hematopoietic stem cell transplantation. Leuk Lymphoma 2017; 59:590-600. [DOI: 10.1080/10428194.2017.1344844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Federico Simonetta
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | | | - Yan Beauverd
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | - Olga Tsopra
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | - Yordanka Tirefort
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | - Aikaterini Koutsi
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | - Caroline Stephan
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | | | - Amandine Pradier
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | - Carole Dantin
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | - Marc Ansari
- Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospitals, Geneva, Switzerland
| | - Eddy Roosnek
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| | - Yves Chalandon
- Department of Oncology, Division of Hematology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
31
|
Role of αβ T Cell Depletion in Prevention of Graft versus Host Disease. Biomedicines 2017; 5:biomedicines5030035. [PMID: 28672883 PMCID: PMC5618293 DOI: 10.3390/biomedicines5030035] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/15/2017] [Accepted: 06/18/2017] [Indexed: 12/18/2022] Open
Abstract
Graft versus host disease (GVHD) represents a major complication of allogeneic hematopoietic stem cell transplantation (allo HCT). Graft cellular manipulation has been used to mitigate the risk of GVHD. The αβ T cells are considered the primary culprit for causing GVHD therefore depletion of this T cell subset emerged as a promising cellular manipulation strategy to overcome the human leukocyte antigen (HLA) barrier of haploidentical (haplo) HCT. This approach is also being investigated in HLA-matched HCT. In several studies, αβ T cell depletion HCT has been performed without pharmacologic GVHD prophylaxis, thus unleashing favorable effect of donor’s natural killer cells (NK) and γδ T cells. This article will discuss the evolution of this method in clinical practice and the clinical outcome as described in different clinical trials.
Collapse
|
32
|
Foell J, Pfirstinger B, Rehe K, Wolff D, Holler E, Corbacioglu S. Haploidentical stem cell transplantation with CD3+-/CD19+- depleted peripheral stem cells for patients with advanced stage sickle cell disease and no alternative donor: results of a pilot study. Bone Marrow Transplant 2017; 52:938-940. [DOI: 10.1038/bmt.2017.49] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
33
|
Ex vivo T-cell depletion in allogeneic hematopoietic stem cell transplant: past, present and future. Bone Marrow Transplant 2017; 52:1241-1248. [PMID: 28319073 PMCID: PMC5589981 DOI: 10.1038/bmt.2017.22] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/05/2016] [Accepted: 12/15/2016] [Indexed: 01/06/2023]
Abstract
The most common cause of post-transplant mortality in patients with hematological malignancy is relapse, followed by GvHD, infections, organ toxicity and second malignancy. Immune-mediated complications such as GvHD continue to be challenging, yet amenable to control through manipulation of the T-cell compartment of the donor graft with subsequent immunomodulation after transplant. However, risk of both relapse and infection increase concomitantly with T-cell depletion (TCD) strategies that impair immune recovery. In this review, we discuss the clinical outcome of current and emerging strategies of TCD in allogeneic hematopoietic stem cell transplant that have developed during the modern transplantation era, focusing specifically on ex vivo strategies that target selected T-cell subsets.
Collapse
|
34
|
González-Vicent M, Molina B, Deltoro N, Sevilla J, Vicario JL, Castillo A, Ramirez M, Díaz MÁ. Donor age matters in T-cell depleted haploidentical hematopoietic stem cell transplantation in pediatric patients: Faster immune reconstitution using younger donors. Leuk Res 2017; 57:60-64. [PMID: 28292719 DOI: 10.1016/j.leukres.2017.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 11/25/2022]
Abstract
T-cell depleted (TCD) haploidentical transplantation is increasingly used in paediatric patients with haematological malignancies and donor selection is a challenge. We conclude that a simple criterion such as donor age should be also considered in depleted haploidentical setting because faster immune reconstitution is achieved using younger donors decreasing non-relapse related mortality.
Collapse
Affiliation(s)
| | - Blanca Molina
- Stem Cell Transplant Unit, Hospital Niño Jesus, Madrid, Spain
| | - Natalia Deltoro
- Stem Cell Transplant Unit, Hospital Niño Jesus, Madrid, Spain
| | | | - José Luis Vicario
- Histocompatibility Laboratory, Community Transfusion Center, Madrid, Spain
| | - Ana Castillo
- Oncology Laboratory, Hospital Niño Jesus, Madrid, Spain
| | | | | |
Collapse
|
35
|
Marks DI, Abid MB. A Stem Cell Donor for Every Adult Requiring an Allograft for Acute Lymphoblastic Leukemia? Biol Blood Marrow Transplant 2017; 23:182-183. [DOI: 10.1016/j.bbmt.2016.11.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 11/26/2022]
|
36
|
Selective Depletion of αβ T Cells and B Cells for Human Leukocyte Antigen–Haploidentical Hematopoietic Stem Cell Transplantation. A Three-Year Follow-Up of Procedure Efficiency. Biol Blood Marrow Transplant 2016; 22:2056-2064. [DOI: 10.1016/j.bbmt.2016.08.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/04/2016] [Indexed: 12/16/2022]
|
37
|
Abstract
Hematopoietic Stem Cells Transplantation (HSCT) is an effective treatment for hematological and non-hematological diseases. The main challenge in autologous HSCT is purging of malignant cells to prevent relapse. In allogeneic HSCT graft-versus-host disease (GvHD) and opportunistic infections are frequent complications. Two types of graft manipulation have been introduced: the first one in the autologous context aimed at separating malignant cells from hematopoietic stem cells (HSC), and the second one in allogeneic HSCT aimed at reducing the incidence of GvHD and at accelerating immune reconstitution. Here we describe the manipulations used for cell purging in autologous HSCT or for T Cell Depletion (TCD) and T cell selection in allogeneic HSCT. More complex manipulations, requiring a Good Manufacturing Practice (GMP) facility, are briefly mentioned.
Collapse
|
38
|
Diaz MA, Pérez-Martínez A, Herrero B, Deltoro N, Martinez I, Ramirez M, Abad L, Sevilla J, Merino E, Ruiz J, Vicario JL, Gonzalez-Vicent M. Prognostic factors and outcomes for pediatric patients receiving an haploidentical relative allogeneic transplant using CD3/CD19-depleted grafts. Bone Marrow Transplant 2016; 51:1211-6. [DOI: 10.1038/bmt.2016.101] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 03/07/2016] [Accepted: 03/11/2016] [Indexed: 11/09/2022]
|
39
|
Granzin M, Soltenborn S, Müller S, Kollet J, Berg M, Cerwenka A, Childs RW, Huppert V. Fully automated expansion and activation of clinical-grade natural killer cells for adoptive immunotherapy. Cytotherapy 2016; 17:621-32. [PMID: 25881519 PMCID: PMC8725994 DOI: 10.1016/j.jcyt.2015.03.611] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 02/28/2015] [Accepted: 03/10/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND AIMS Ex vivo expansion of natural killer (NK) cells is a strategy to produce large numbers of these effector cells for immunotherapy. However, the transfer of bench-top expansion protocols to clinically applicable methods is challenging for NK cell-based therapy because of regulatory aspects and scale-up issues. Therefore, we developed an automated, large-scale NK cell expansion process. METHODS Enriched NK cells were expanded with interleukin-2 and irradiated clinical-grade Epstein-Barr virus-transformed lymphoblastoid feeder cells with the use of an automated system in comparison to manual expansion, and the cells were investigated for their functionality, phenotype and gene expression. RESULTS Automated expansion resulted in a mean 850-fold expansion of NK cells by day 14, yielding 1.3 (± 0.9) × 10(9) activated NK cells. Automatically and manually produced NK cells were comparable in target cell lysis, degranulation and production of interferon-γ and tumor necrosis factor-α and had similar high levels of antibody-dependent cellular cytotoxicity against rituximab-treated leukemic cells. NK cells after automated or manual expansion showed similar gene expression and marker profiles. However, expanded NK cells differed significantly from primary NK cells including upregulation of the functional relevant molecules TRAIL and FasL and NK cell-activating receptors NKp30, NKG2D and DNAM-1. Neither automatically nor manually expanded NK cells showed reduced telomere length indicative of a conserved proliferative potential. CONCLUSIONS We established an automated method to expand high numbers of clinical-grade NK cells with properties similar to their manually produced counterparts. This automated process represents a highly efficient tool to standardize NK cell processing for therapeutic applications.
Collapse
Affiliation(s)
- Markus Granzin
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany; Innate Immunity Group, German Cancer Research Center, Heidelberg, Germany.
| | | | | | - Jutta Kollet
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Maria Berg
- National Heart, Lung and Blood Institute, Hematology Branch, Bethesda, Maryland, USA
| | - Adelheid Cerwenka
- Innate Immunity Group, German Cancer Research Center, Heidelberg, Germany
| | - Richard W Childs
- National Heart, Lung and Blood Institute, Hematology Branch, Bethesda, Maryland, USA
| | | |
Collapse
|
40
|
Avecilla ST, Goss C, Bleau S, Tonon JA, Meagher RC. How do I perform hematopoietic progenitor cell selection? Transfusion 2016; 56:1008-12. [PMID: 26919388 DOI: 10.1111/trf.13534] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/13/2016] [Accepted: 01/13/2016] [Indexed: 12/14/2022]
Abstract
Graft-versus-host disease remains the most important source of morbidity and mortality associated with allogeneic stem cell transplantation. The implementation of hematopoietic progenitor cell (HPC) selection is employed by some stem cell processing facilities to mitigate this complication. Current cell selection methods include reducing the number of unwanted T cells (negative selection) and/or enriching CD34+ hematopoietic stem/progenitors (positive selection) using immunomagnetic beads subjected to magnetic fields within columns to separate out targeted cells. Unwanted side effects of cell selection as a result of T-cell reduction are primary graft failure, increased infection rates, delayed immune reconstitution, possible disease relapse, and posttransplant lymphoproliferative disease. The Miltenyi CliniMACS cell isolation system is the only device currently approved for clinical use by the Food and Drug Administration. It uses magnetic microbeads conjugated with a high-affinity anti-CD34 monoclonal antibody capable of binding to HPCs in marrow, peripheral blood, or umbilical cord blood products. The system results in significantly improved CD34+ cell recoveries (50%-100%) and consistent 3-log CD3+ T-cell reductions compared to previous generations of CD34+ cell selection procedures. In this article, the CliniMACS procedure is described in greater detail and the authors provide useful insight into modifications of the system. Successful implementation of cell selection procedures can have a significant positive clinical effect by greatly increasing the pool of donors for recipients requiring transplants. However, before a program implements cell selection techniques, it is important to consider the time and financial resources required to properly and safely perform these procedures.
Collapse
Affiliation(s)
- Scott T Avecilla
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cheryl Goss
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sharon Bleau
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jo-Ann Tonon
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard C Meagher
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
41
|
Bremm M, Cappel C, Erben S, Jarisch A, Schumm M, Arendt A, Bonig H, Klingebiel T, Koehl U, Bader P, Huenecke S. Generation and flow cytometric quality control of clinical-scale TCRαβ/CD19-depleted grafts. CYTOMETRY PART B-CLINICAL CYTOMETRY 2015; 92:126-135. [DOI: 10.1002/cyto.b.21328] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/16/2015] [Accepted: 09/17/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Melanie Bremm
- Clinic for Pediatric and Adolescent Medicine; University Hospital; Frankfurt Germany
| | - Claudia Cappel
- Clinic for Pediatric and Adolescent Medicine; University Hospital; Frankfurt Germany
| | - Stephanie Erben
- Clinic for Pediatric and Adolescent Medicine; University Hospital; Frankfurt Germany
| | - Andrea Jarisch
- Clinic for Pediatric and Adolescent Medicine; University Hospital; Frankfurt Germany
| | - Michael Schumm
- Department of Hematology/Oncology; Children's University Hospital; Tübingen Germany
| | | | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt/Main, Division for Cell Processing, German Red Cross Blood Donor Service Baden-Württemberg-Hessen; Frankfurt/Main Germany
| | - Thomas Klingebiel
- Clinic for Pediatric and Adolescent Medicine; University Hospital; Frankfurt Germany
| | - Ulrike Koehl
- Institute of Cellular Therapeutics, GMP Development Unit; IFB-TX; Hannover Medical School; Hannover Germany
| | - Peter Bader
- Clinic for Pediatric and Adolescent Medicine; University Hospital; Frankfurt Germany
| | - Sabine Huenecke
- Clinic for Pediatric and Adolescent Medicine; University Hospital; Frankfurt Germany
| |
Collapse
|
42
|
Falkenburg JHF, Jedema I. Allo-reactive T cells for the treatment of hematological malignancies. Mol Oncol 2015; 9:1894-903. [PMID: 26578450 DOI: 10.1016/j.molonc.2015.10.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 12/11/2022] Open
Abstract
Several mechanisms can be responsible for control of hematological tumors by allo-reactive T cells. Following allogeneic stem cell transplantation (alloSCT) donor T cells recognizing genetic disparities presented on recipient cells and not on donor cells are main effectors of tumor control, but also of the detrimental graft versus host disease (GVHD). Since after transplantation normal hematopoiesis is of donor origin, any T cell response directed against polymorphic antigens expressed on hematopoietic recipient cells but not on donor cells will result in an anti-tumor response not affecting normal hematopoiesis. After fully HLA-matched alloSCT, T cells recognizing polymorphic peptides derived from proteins encoded by genes selectively expressed in hematopoietic lineages may result in anti-tumor responses without GVHD. Due to the high susceptibility of hematopoietic cells for T cell recognition, a low amplitude of the overall T cell response may also be in favor of the anti-tumor reactivity in hematological malignancies. A mismatch between donor and patient for specific HLA-alleles can also be exploited to induce a selective T cell response against patient (malignant) hematopoietic cells. If restricting HLA class II molecules are selectively expressed on hematopoietic cells under non-inflammatory circumstances, allo HLA class-II responses may control the tumor with limited risk of GVHD. Alternatively, T cells recognizing hematopoiesis-restricted antigens presented in the context of mismatched HLA alleles may be used to treat patients with hematological cancers. This review discusses various ways to manipulate the allo-immune response aiming to exploit the powerful ability of allo-reactive T-cells to control the malignancies without causing severe damage to non-hematopoietic tissues.
Collapse
Affiliation(s)
- J H F Falkenburg
- Department of Hematology, Leiden University Medical Center, Netherlands
| | - I Jedema
- Department of Hematology, Leiden University Medical Center, Netherlands.
| |
Collapse
|
43
|
Balashov D, Shcherbina A, Maschan M, Trakhtman P, Skvortsova Y, Shelikhova L, Laberko A, Livshits A, Novichkova G, Maschan A. Single-Center Experience of Unrelated and Haploidentical Stem Cell Transplantation with TCRαβ and CD19 Depletion in Children with Primary Immunodeficiency Syndromes. Biol Blood Marrow Transplant 2015; 21:1955-62. [PMID: 26187864 DOI: 10.1016/j.bbmt.2015.07.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/08/2015] [Indexed: 10/23/2022]
Abstract
The transplantation of stem cells from a matched unrelated donor (MUD) or a haploidentical mismatched related donor (MMRD) is a widely used variant of curative treatment for patients with primary immunodeficiency (PID). Currently, different strategies are used to reduce the risk of post-transplant complications and enhance immune reconstitution. We report the preliminary results of MUD and MMRD transplantation with TCRαβ/CD19 depletion in patients with PID (trial registered at www.clinicaltrials.gov as NCT02327351). Thirty-seven PID patients (median age, 2.6 years; range, .2 to 17) were transplanted from MUDs (n = 27) or haploidentical MMRDs (n = 10) after TCRαβ(+)/CD19(+) graft depletion. The median numbers of CD34(+) and TCRαβ(+) cells in the graft were 11.7 × 10(6)/kg and 10.6 × 10(3)/kg, respectively. Acute graft-versus-host disease (GVHD) was observed in 8 patients (22%), without a statistically significant difference between MUDs and MMRDs; 7 of these patients had grade II acute GVHD and responded to first-line therapy, whereas 1 patient had grade IV acute GVHD with transformation to extensive chronic GVHD. Primary and secondary graft failure (nonengraftment or rejection) was observed in 10 patients (27%), 9 of whom were treated with 1 alkylating agent in the conditioning regimen. All these patients were successfully retransplanted with different rescue protocols. Preliminary data on immune reconstitution were very encouraging. Most patients had significant numbers of T lymphocytes detected on the first assessment (day +30) and more than 500 T cells/μL, on day +120. Based on our preliminary data, no significant difference was seen between MMRD and MUD hematopoietic stem cell transplantation (HSCT). With a median follow-up period of 15 months, the cumulative probabilities of overall patient survival and transplant-related mortality were 96.7% and 3.3%, respectively. Based on the results, the ability to control the main post-transplant complications and the immune reconstitution rates are the main factors leading to successful outcome in patients with PID after TCRαβ(+)-depleted HSCT.
Collapse
Affiliation(s)
- Dmitry Balashov
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| | - Anna Shcherbina
- Department of Clinical Immunology, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Michael Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Pavel Trakhtman
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Yulia Skvortsova
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Larisa Shelikhova
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexandra Laberko
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Anna Livshits
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Galina Novichkova
- Department of pediatric hematology and oncology, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexei Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev Federal Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
44
|
de Witte MA, Kierkels GJJ, Straetemans T, Britten CM, Kuball J. Orchestrating an immune response against cancer with engineered immune cells expressing αβTCRs, CARs, and innate immune receptors: an immunological and regulatory challenge. Cancer Immunol Immunother 2015; 64:893-902. [PMID: 25990073 PMCID: PMC4481298 DOI: 10.1007/s00262-015-1710-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 05/04/2015] [Indexed: 12/20/2022]
Abstract
Over half a century ago, the first allogeneic stem cell transplantation (allo-SCT) initiated cellular immunotherapy. For several decades, little progress was made, and toxicity of allo-SCT remained a major challenge. However, recent breakthroughs have opened new avenues to further develop this modality and to provide less toxic and equally efficient interventions for patients suffering from hematological or solid malignancies. Current novel cellular immune interventions include ex vivo expansion and adoptive transfer of tumor-infiltrating immune cells or administration of drugs which antagonize tolerizing mechanisms. Alternatively, transfer of immune cells engineered to express defined T cell receptors (TCRs) and chimeric antigen receptors (CARs) has shown its potential. A valuable addition to ‘engineered’ adaptive immunity has emerged recently through the improved understanding of how innate immune cells can attack cancer cells without substantial side effects. This has enabled the development of transplantation platforms with limited side effects allowing early immune interventions as well as the design of engineered immune cells expressing innate immune receptors. Here, we focus on innate immune interventions and their orchestration with TCR- and CAR-engineered immune cells. In addition, we discuss how the exploitation of the full potential of cellular immune interventions is influenced by regulatory frameworks. Finally, we highlight and discuss substantial differences in the current landscape of clinical trials in Europe as compared to the USA. The aim is to stimulate international efforts to support regulatory authorities and funding agencies, especially in Europe, to create an environment that will endorse the development of engineered immune cells for the benefit of patients.
Collapse
Affiliation(s)
- Moniek A de Witte
- Department of Hematology, University Medical Center Utrecht, Room Number Q05.4.301, PO Box 85500, 3508, GA, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
45
|
Rapid memory T-cell reconstitution recapitulating CD45RA-depleted haploidentical transplant graft content in patients with hematologic malignancies. Bone Marrow Transplant 2015; 50:968-77. [PMID: 25665048 PMCID: PMC4636007 DOI: 10.1038/bmt.2014.324] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/18/2014] [Accepted: 12/12/2014] [Indexed: 12/20/2022]
Abstract
T-cell depletion of an HLA-haploidentical graft is often used to prevent graft-vs.-host disease (GvHD), but the procedure may lead to increased graft failure, relapse, and infections due to delayed immune recovery. We hypothesized that selective depletion of the CD45RA+ subset can effectively reduce GvHD through removal of naïve T cells, while providing improved donor immune reconstitution through adoptive transfer of CD45RA– memory T cells. Herein, we present results from the first 17 patients with poor-prognosis hematologic malignancy who received haploidentical donor transplantation with CD45RA-depleted progenitor cell grafts following a novel reduced intensity conditioning regimen without total body irradiation or serotherapy. Extensive depletion of CD45RA+ T cells and B cells, with preservation of abundant memory T cells, was consistently achieved in all 17 products. Neutrophil engraftment (median day +10) and full donor chimerism (median day +11) was rapidly achieved post-transplantation. Early T-cell reconstitution directly correlated with the CD45RA-depleted graft content. T-cell function recovered rapidly with broad TCR Vβ spectra. There was no infection-related mortality in this heavily pretreated population, and no patient developed acute GvHD despite infusion of a median of >100 million per kilogram haploidentical T cells.
Collapse
|
46
|
Control of murine cytomegalovirus infection by γδ T cells. PLoS Pathog 2015; 11:e1004481. [PMID: 25658831 PMCID: PMC4450058 DOI: 10.1371/journal.ppat.1004481] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 09/18/2014] [Indexed: 12/28/2022] Open
Abstract
Infections with cytomegalovirus (CMV) can cause severe disease in immunosuppressed patients and infected newborns. Innate as well as cellular and humoral adaptive immune effector functions contribute to the control of CMV in immunocompetent individuals. None of the innate or adaptive immune functions are essential for virus control, however. Expansion of γδ T cells has been observed during human CMV (HCMV) infection in the fetus and in transplant patients with HCMV reactivation but the protective function of γδ T cells under these conditions remains unclear. Here we show for murine CMV (MCMV) infections that mice that lack CD8 and CD4 αβ-T cells as well as B lymphocytes can control a MCMV infection that is lethal in RAG-1-/- mice lacking any T- and B-cells. γδ T cells, isolated from infected mice can kill MCMV infected target cells in vitro and, importantly, provide long-term protection in infected RAG-1-/- mice after adoptive transfer. γδ T cells in MCMV infected hosts undergo a prominent and long-lasting phenotypic change most compatible with the view that the majority of the γδ T cell population persists in an effector/memory state even after resolution of the acute phase of the infection. A clonotypically focused Vγ1 and Vγ2 repertoire was observed at later stages of the infection in the organs where MCMV persists. These findings add γδ T cells as yet another protective component to the anti-CMV immune response. Our data provide clear evidence that γδ T cells can provide an effective control mechanism of acute CMV infections, particularly when conventional adaptive immune mechanisms are insufficient or absent, like in transplant patient or in the developing immune system in utero. The findings have implications in the stem cell transplant setting, as antigen recognition by γδ T cells is not MHC-restricted and dual reactivity against CMV and tumors has been described. Cytomegalovirus is a clinically important pathogen. While infection in hosts with a functional immune system is usually asymptomatic, the virus can cause significant morbidity and mortality in individuals with an immature or suppressed immune system. The virus causes severe clinical complication in transplant recipients and congenital CMV infections are the most common infectious cause of neurological disorders in children. Multiple layers of innate and adoptive immunity are involved in the control of CMV and single deficiencies of one immune cell type can be compensated by other immune cells. Expansions of γδ T lymphocytes, which are regarded as innate-like cells with adaptive-like potential, have been shown to be associated with CMV infections in human transplant patients and neonates. Their role in protective immunity against CMV has been unclear, however. Here we show direct evidence in the murine CMV model (MCMV) that γδ T lymphocytes can provide protection against a lethal MCMV infection in the absence of any other cells of the adoptive immune system. Upon infection, γδ T lymphocytes undergo a significant expansion and a prominent and long-lasting phenotypic change. These findings have implications for the development of new cellular therapy regimens in CMV infections in the transplant setting that should be evaluated in the future.
Collapse
|
47
|
Seidel UJE, Vogt F, Grosse-Hovest L, Jung G, Handgretinger R, Lang P. γδ T Cell-Mediated Antibody-Dependent Cellular Cytotoxicity with CD19 Antibodies Assessed by an Impedance-Based Label-Free Real-Time Cytotoxicity Assay. Front Immunol 2014; 5:618. [PMID: 25520723 PMCID: PMC4251440 DOI: 10.3389/fimmu.2014.00618] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/18/2014] [Indexed: 11/13/2022] Open
Abstract
γδ T cells are not MHC restricted, elicit cytotoxicity against various malignancies, are present in early post-transplant phases in novel stem cell transplantation strategies and have been shown to mediate antibody-dependent cellular cytotoxicity (ADCC) with monoclonal antibodies (mAbs). These features make γδ T cells promising effector cells for antibody-based immunotherapy in pediatric patients with B-lineage acute lymphoblastic leukemia (ALL). To evaluate combination of human γδ T cells with CD19 antibodies for immunotherapy of B-lineage ALL, γδ T cells were expanded after a GMP-compliant protocol and ADCC of both primary and expanded γδ T cells with an Fc-optimized CD19 antibody (4G7SDIE) and a bi-specific antibody with the specificities CD19 and CD16 (N19-C16) was evaluated in CD107a-degranulation assays and intracellular cytokine staining. CD107a, TNFα, and IFNγ expression of primary γδ T cells were significantly increased and correlated with CD16-expression of γδ T cells. γδ T cells highly expressed CD107a after expansion and no further increased expression by 4G7SDIE and N19-C16 was measured. Cytotoxicity of purified expanded γδ T cells targeting CD19-expressing cells was assessed in both europium-TDA release and in an impedance-based label-free method (using the xCELLigence system) measuring γδ T cell lysis in real-time. Albeit in the 2 h end-point europium-TDA release assay no increased lysis was observed, in real-time xCELLigence assays both significant antibody-independent cytotoxicity and ADCC of γδ T cells were observed. The xCELLigence system outperformed the end-point europium-TDA release assay in sensitivity and allows drawing of conclusions to lysis kinetics of γδ T cells over prolonged periods of time periods. Combination of CD19 antibodies with primary as well as expanded γδ T cells exhibits a promising approach, which may enhance clinical outcome of patients with pediatric B-lineage ALL and requires clinical evaluation.
Collapse
Affiliation(s)
- Ursula Jördis Eva Seidel
- Department of General Paediatrics, Oncology/Haematology, University Children's Hospital Tübingen , Tübingen , Germany
| | - Fabian Vogt
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen , Tübingen , Germany ; Partner Site Tübingen, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) , Tübingen , Germany
| | - Ludger Grosse-Hovest
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen , Tübingen , Germany ; SYNIMMUNE GmbH , Tübingen , Germany
| | - Gundram Jung
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen , Tübingen , Germany ; Partner Site Tübingen, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) , Tübingen , Germany
| | - Rupert Handgretinger
- Department of General Paediatrics, Oncology/Haematology, University Children's Hospital Tübingen , Tübingen , Germany ; Partner Site Tübingen, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) , Tübingen , Germany
| | - Peter Lang
- Department of General Paediatrics, Oncology/Haematology, University Children's Hospital Tübingen , Tübingen , Germany ; Partner Site Tübingen, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) , Tübingen , Germany
| |
Collapse
|
48
|
Schwinger W, Sovinz P, Benesch M, Lackner H, Seidel M, Strenger V, Sperl D, Raicht A, Brunner-Krainz M, Paschke E, Plecko B, Urban C. Unrelated CD3/CD19-depleted peripheral stem cell transplantation for Hurler syndrome. Pediatr Hematol Oncol 2014; 31:723-30. [PMID: 25116402 DOI: 10.3109/08880018.2014.939794] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
For patients with mucopolysaccharidosis type IH (MPS1-H; Hurler syndrome), early allogeneic hematopoietic stem cell transplantation (HSCT) is the treatment of choice. One boy and one girl aged 20.5 and 22 months, respectively, with MPS1-H received a conditioning regimen consisting of thiotepa, fludarabine, treosulfan, and ATG. Grafts were peripheral blood stem cells from unrelated donors (10/12 and 11/11 matched), that were manipulated by CD3/CD19 depletion and contained 20.3 and 28.2 × 10(6) CD34+ cells/kg body weight, respectively. Both patients achieved stable hematopoietic engraftment and stable donor chimerism. Neither acute or chronic graft-versus-host disease (GVHD) nor other severe transplant-related complications occurred. At a follow-up of 48 and 37 months, both patients are alive and well with normal levels of α-L-iduronidase and have made major neurodevelopmental progress. Treosulfan-based conditioning offers the advantage of reduced toxicity; the use of unrelated CD3/CD19-depleted peripheral stem cell grafts allows transfusion of high CD34+ cell numbers together with a "tailored" number of CD3+ cells as well as engraftment facilitating cells in order to achieve rapid hematopoietic engraftment while reducing the risk of graft rejection and GVHD. This regimen might be an additional option when unrelated donor HSCT is considered for a patient with MPS1-H.
Collapse
Affiliation(s)
- Wolfgang Schwinger
- 1Division of Pediatric Hematology/Oncology, University Children's Hospital, Graz, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Chang YJ, Wang Y, Huang XJ. Haploidentical stem cell transplantation for the treatment of leukemia: current status. Expert Rev Hematol 2014; 7:635-47. [DOI: 10.1586/17474086.2014.954543] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Abstract
Over the past 5 years, many novel approaches to early diagnosis, prevention, and treatment of acute graft-versus-host disease (aGVHD) have been translated from the bench to the bedside. In this review, we highlight recent discoveries in the context of current aGVHD care. The most significant innovations that have already reached the clinic are prophylaxis strategies based upon a refinement of our understanding of key sensors, effectors, suppressors of the immune alloreactive response, and the resultant tissue damage from the aGVHD inflammatory cascade. In the near future, aGVHD prevention and treatment will likely involve multiple modalities, including small molecules regulating immunologic checkpoints, enhancement of suppressor cytokines and cellular subsets, modulation of the microbiota, graft manipulation, and other donor-based prophylaxis strategies. Despite long-term efforts, major challenges in treatment of established aGVHD still remain. Resolution of inflammation and facilitation of rapid immune reconstitution in those with only a limited response to corticosteroids is a research arena that remains rife with opportunity and urgent clinical need.
Collapse
|