1
|
Wiercinska E, Seifried E, Bonig H. CD3/CD19 Depletion for T-cell Reduction of Allogeneic Transplants: Mostly Efficient, but not Robust. Clin Hematol Int 2021; 3:103-107. [PMID: 34820615 PMCID: PMC8486974 DOI: 10.2991/chi.k.210725.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Aggressive T-cell depletion, in vitro or in vivo, is a prerequisite for survival of haplo-identical stem cell transplantation. The classical T-cell-depleted transplant, immunomagnetically enriched CD34+ cells, is very safe with respect to graft-versus-host reactivity, but associated with very high transplant-related and relapse mortality with an overall probability of survival of only 20%. Protocols for T- and B-cell depletion were therefore developed, reasoning that transplantation of the majority of Natural Killer (NK) cells and the substantial dose of residual T-cells might improve survival, which was, in principle, confirmed. Anecdotal reports of frequent failure to achieve adequate T-cell depletion prompted review of the aggregate data for transplant quality at our center. The first observation is the relative paucity of combined CD3/CD19 depletion processes as PTCy protocols have made inroads, 13 depletions in 8 years. Median T- and B-cell log-depletion were -3.89 and -1.92, respectively; instead of, CD34+ cell recovery was generally high (median 92%), as was NK-cell recovery (median 52%). However, the process failed to yield satisfactory T- and B-cell depletion in two out of 13 preparations, of which one product could be rescued by a second round of depletion, at the expense of CD34+ cell recovery. In our hands, the process is thus insufficiently robust for routine clinical use. Assuming similar observations in other centers, this may explain implementation of alternative protocols, such as TCRαβ/CD19 depletion or transplantation of unmanipulated grafts with subsequent in vivo depletion.
Collapse
Affiliation(s)
- Eliza Wiercinska
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt a.M., Germany
| | - Erhard Seifried
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt a.M., Germany.,Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt a.M., Germany
| | - Halvard Bonig
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt a.M., Germany.,Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt a.M., Germany.,Department of Medicine/Division of Hematology, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Bremm M, Krastel T, Cappel C, Zimmermann O, Pfeffermann LM, Katzki V, Bonig H, Schäfer R, Rettinger E, Merker M, Bremm S, Schaefer K, Klingebiel T, Soerensen J, Bader P, Huenecke S. Depletion of CD45RA + T cells: Advantages and disadvantages of different purification methods. J Immunol Methods 2021; 492:112960. [PMID: 33417916 DOI: 10.1016/j.jim.2021.112960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 12/03/2020] [Accepted: 12/31/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Recently, new advances were made regarding the depletion of CD45RA+ naïve T cells from haploidentical grafts as they are suspected to be the most alloreactive. METHODS Within this project we investigated CD45RA-depletion from G-CSF mobilized PBSC by two different purification strategies according to GMP, specifically direct depletion of CD45RA+ cells (one-step approach), or CD34-positive selection followed by CD45RA-depletion (two-step approach). RESULTS With log -3.9 and - 3.8 the depletion quality of CD45RA+ T cells was equally for both approaches together with a close to complete CD19+ B cell depletion. However, due to a high expression of CD45RA the majority of NK cells were lost within both CD45RA depletion strategies. Stem cell recovery after one-step CD45RA-depletion was at median 52.0% (range: 49.7-67.2%), which was comparable to previously published recovery data received from direct CD34 positive selection. Memory T cell recovery including CD4+ and CD8+ memory T cell subsets was statistically not differing between both purification approaches. The recovery of CD4+ and CD8+ T cells was as well similar, but overall a higher amount of cytotoxic than T-helper cells were lost as indicated by an increase of the CD4/CD8 ratio. CONCLUSIONS CD45RA-depletion from G-CSF mobilized PBSC is feasible as one- and two-step approach and results in sufficient reduction of CD45RA+ T cells as well as B cells, but also to a co-depletion of NK cells. However, by gaining two independent cell products, the two-step approach enables the highest clinical flexibility in regard to individual graft composition with precise dosage of stem cells and T cells.
Collapse
Affiliation(s)
- Melanie Bremm
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany.
| | - Theresa Krastel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Claudia Cappel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Olga Zimmermann
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Lisa-Marie Pfeffermann
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Verena Katzki
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt/Main, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Frankfurt/Main, Germany
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt/Main, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Frankfurt/Main, Germany
| | - Eva Rettinger
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Michael Merker
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Sebastian Bremm
- Data Analytics & Visualization, Frankfurt University of Applied Sciences, Frankfurt/Main, Germany
| | - Kirsten Schaefer
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Thomas Klingebiel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Jan Soerensen
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Peter Bader
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| | - Sabine Huenecke
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt/Main, Germany
| |
Collapse
|
3
|
Heinze A, Grebe B, Bremm M, Huenecke S, Munir TA, Graafen L, Frueh JT, Merker M, Rettinger E, Soerensen J, Klingebiel T, Bader P, Ullrich E, Cappel C. The Synergistic Use of IL-15 and IL-21 for the Generation of NK Cells From CD3/CD19-Depleted Grafts Improves Their ex vivo Expansion and Cytotoxic Potential Against Neuroblastoma: Perspective for Optimized Immunotherapy Post Haploidentical Stem Cell Transplantation. Front Immunol 2019; 10:2816. [PMID: 31849984 PMCID: PMC6901699 DOI: 10.3389/fimmu.2019.02816] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/15/2019] [Indexed: 12/21/2022] Open
Abstract
Neuroblastoma (NB) is the most common solid extracranial tumor in childhood. Despite therapeutic progress, prognosis in high-risk NB is poor and innovative therapies are urgently needed. Therefore, we addressed the potential cytotoxic capacity of interleukin (IL)-activated natural killer (NK) cells compared to cytokine-induced killer (CIK) cells for the treatment of NB. NK cells were isolated from peripheral blood mononuclear cells (PBMCs) by indirect CD56-enrichment or CD3/CD19-depletion and expanded with different cytokine combinations, such as IL-2, IL-15, and/or IL-21 under feeder-cell free conditions. CIK cells were generated from PBMCs by ex vivo stimulation with interferon-γ, IL-2, OKT-3, and IL-15. Comparative analysis of expansion rate, purity, phenotype and cytotoxicity was performed. CD56-enriched NK cells showed a median expansion rate of 4.3-fold with up to 99% NK cell content. The cell product after CD3/CD19-depletion consisted of a median 43.5% NK cells that expanded significantly faster reaching also 99% of NK cell purity. After 10–12 days of expansion, both NK cell preparations showed a significantly higher median cytotoxic capacity against NB cells relative to CIK cells. Remarkably, these NK cells were also capable of efficiently killing NB spheroidal 3D culture in long-term cytotoxicity assays. Further optimization using a novel NK cell culture medium and a prolonged culturing procedure after CD3/CD19-depletion for up to 15 days enhanced the expansion rate up to 24.4-fold by maintaining the cytotoxic potential. Addition of an IL-21 boost prior to harvesting significantly increased the cytotoxicity. The final cell product consisted for the major part of CD16−, NCR-expressing, poly-functional NK cells with regard to cytokine production, CD107a degranulation and antitumor capacity. In summary, our study revealed that NK cells have a significantly higher cytotoxic potential to combat NB than CIK cell products, especially following the synergistic use of IL-15 and IL-21 for NK cell activation. Therefore, the use of IL-15+IL-21 expanded NK cells generated from CD3/CD19-depleted apheresis products seems to be highly promising as an immunotherapy in combination with haploidentical stem cell transplantation (SCT) for high-risk NB patients.
Collapse
Affiliation(s)
- Annekathrin Heinze
- Experimental Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Beatrice Grebe
- Experimental Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Melanie Bremm
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Sabine Huenecke
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Tasleem Ah Munir
- Experimental Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Lea Graafen
- Experimental Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Jochen T Frueh
- Experimental Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Michael Merker
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Eva Rettinger
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Jan Soerensen
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Thomas Klingebiel
- Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Evelyn Ullrich
- Experimental Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt am Main, Frankfurt am Main, Germany
| | - Claudia Cappel
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
4
|
Yoon EJ, Zhang J, Weinberg RS, Brochstein JA, Nandi V, Sachais BS, Shi PA. Validation of simple prediction algorithms to consistently achieve CD3+ and postselection CD34+ targets with leukapheresis. Transfusion 2019; 60:133-143. [PMID: 31756000 DOI: 10.1111/trf.15576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cellular therapies using engineered T cells, haploidentical transplants, and autologous gene therapy are increasing. Specified CD3+ or high CD34+ doses are typically required for subsequent manufacturing, manipulation, or CD34+ selection. Simple, practical, and reliable lymphocyte and hematopoietic progenitor cell (HPC) collection algorithms accounting for subsequent CD34+ selection have not been published. STUDY DESIGN AND METHODS In this analysis of 15 haploidentical donors undergoing tandem lymphocyte and HPC collections, we validated one-step, practical prediction algorithms (Appendix S1, available as supporting information in the online version of this paper) that use conservative facility-specific collection efficiencies, CD34+ selection efficiency, and donor-specific peripheral counts to reliably achieve the target CD3+ and CD34+ product doses. These algorithms expand on our previously published work regarding predictive HPC collection algorithms. RESULTS Ninety-three percent of lymphocyte and 93% of CD34+ collections achieved the final target CD3+ and CD34+ product dose when our algorithm-calculated process volumes were used. Linear regression analysis of our algorithms for CD3+, preselection CD34+, and postselection CD34+ showed statistically significant models with R2 of 0.80 (root mean square error [RMSE], 31.3), 0.72 (RMSE, 385.7), and 0.56 (RMSE, 326.0), respectively, all with p values less than 0.001. CONCLUSION Because achievement of CD3+ or CD34+ dose targets may be critical for safety and efficacy of cell therapies, these simple, practical, and reliable prediction algorithms for lymphocyte and HPC collections should be very useful for collection facilities.
Collapse
Affiliation(s)
- Edward J Yoon
- Clinical Services, New York Blood Center, New York, New York.,Temple University Hospital, Philadelphia, Pennsylvania
| | - Jiahao Zhang
- Clinical Services, New York Blood Center, New York, New York
| | - Rona S Weinberg
- Clinical Services, New York Blood Center, New York, New York.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York
| | - Joel A Brochstein
- Division of Pediatric Hematology-Oncology, Northwell Health, New Hyde Park, New York
| | - Vijay Nandi
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York
| | - Bruce S Sachais
- Clinical Services, New York Blood Center, New York, New York.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York
| | - Patricia A Shi
- Clinical Services, New York Blood Center, New York, New York.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York
| |
Collapse
|
5
|
Singh J, Zúñiga-Pflücker JC. Producing proT cells to promote immunotherapies. Int Immunol 2019; 30:541-550. [PMID: 30102361 DOI: 10.1093/intimm/dxy051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 08/08/2018] [Indexed: 12/17/2022] Open
Abstract
T lymphocytes are critical mediators of the adaptive immune system and they can be harnessed as therapeutic agents against pathogens and in cancer immunotherapy. T cells can be isolated and expanded from patients and potentially generated in vitro using clinically relevant systems. An ultimate goal for T-cell immunotherapy is to establish a safe, universal effector cell type capable of transcending allogeneic and histocompatibility barriers. To this end, human pluripotent stem cells offer an advantage in generating a boundless supply of T cells that can be readily genetically engineered. Here, we review emerging T-cell therapeutics, including tumor-infiltrating lymphocytes, chimeric antigen receptors and progenitor T cells (proT cells) as well as feeder cell-free in vitro systems for their generation. Furthermore, we explore their potential for adoption in the clinic and highlight the challenges that must be addressed to increase the therapeutic success of a universal immunotherapy.
Collapse
Affiliation(s)
- Jastaranpreet Singh
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | |
Collapse
|
6
|
Haase A, Glienke W, Engels L, Göhring G, Esser R, Arseniev L, Martin U. GMP-compatible manufacturing of three iPS cell lines from human peripheral blood. Stem Cell Res 2019; 35:101394. [PMID: 30772682 DOI: 10.1016/j.scr.2019.101394] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/02/2019] [Accepted: 01/26/2019] [Indexed: 10/27/2022] Open
Abstract
The utilization of human induced pluripotent stem cells (hiPSCs) for disease modeling and drug discovery is already reality, and several first-in-man-applications as cellular therapeutics have been initiated. Implementation of good manufacturing practice (GMP)-compliant protocols for the generation of hiPSC lines is crucial to increase the application safety as well as to fulfil the legal requirements for clinical trials approval. Here we describe the development of a GMP-compatible protocol for the reprogramming of CD34+ hematopoietic stem cells from peripheral blood (CD34+ PBHSC) into hiPSCs using Sendai virus-based reprogramming vectors. Three GMP-compatible hiPSC (GMP-hiPSC) lines were manufactured and characterized under these conditions.
Collapse
Affiliation(s)
- Alexandra Haase
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), REBIRTH Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Wolfgang Glienke
- GMP Development Unit, Institute of Cellular Therapeutics, Integrated Research and Treatment Center, Hannover Medical School, Germany
| | - Lena Engels
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), REBIRTH Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Gudrun Göhring
- Institute for Human Genetics, Hannover Medical School, Germany
| | - Ruth Esser
- GMP Development Unit, Institute of Cellular Therapeutics, Integrated Research and Treatment Center, Hannover Medical School, Germany
| | - Lubomir Arseniev
- GMP Development Unit, Institute of Cellular Therapeutics, Integrated Research and Treatment Center, Hannover Medical School, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), REBIRTH Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
7
|
[What is established in cell therapies? : Possibilities and limits in immuno-oncology]. Internist (Berl) 2018; 59:1230-1238. [PMID: 30367191 DOI: 10.1007/s00108-018-0516-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cell and gene therapy as part of immuno-oncology has reached an important milestone in medicine. After decades of experience stem cell transplantation is well established with worldwide >1 million transplantations to date. Due to the improved success of the last years using chimeric antigen receptor (CAR) T cells for CD19 positive leukemia and lymphomas, the interest in cellular therapies is continuously increasing. The current review also gives a short overview about donor lymphocytes, antigen-specific T cells, regulatory T cells, natural killer (NK) cells, mesenchymal stromal cells and induced pluripotent stem (iPS) cells in immuno-oncology.
Collapse
|
8
|
Unrelated donor vs HLA-haploidentical α/β T-cell- and B-cell-depleted HSCT in children with acute leukemia. Blood 2018; 132:2594-2607. [PMID: 30348653 DOI: 10.1182/blood-2018-07-861575] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/03/2018] [Indexed: 12/13/2022] Open
Abstract
Traditionally, hematopoietic stem cell transplantation (HSCT) from both HLA-matched related and unrelated donors (UD) has been used for treating children with acute leukemia (AL) in need of an allograft. Recently, HLA-haploidentical HSCT after αβ T-cell/B-cell depletion (αβhaplo-HSCT) was shown to be effective in single-center studies. Here, we report the first multicenter retrospective analysis of 127 matched UD (MUD), 118 mismatched UD (MMUD), and 98 αβhaplo-HSCT recipients, transplanted between 2010 and 2015, in 13 Italian centers. All these AL children were transplanted in morphological remission after a myeloablative conditioning regimen. Graft failure occurred in 2% each of UD-HSCT and αβhaplo-HSCT groups. In MUD vs MMUD-HSCT recipients, the cumulative incidence of grade II to IV and grade III to IV acute graft-versus-host disease (GVHD) was 35% vs 44% and 6% vs 18%, respectively, compared with 16% and 0% in αβhaplo-HSCT recipients (P < .001). Children treated with αβhaplo-HSCT also had a significantly lower incidence of overall and extensive chronic GVHD (P < .01). Eight (6%) MUD, 32 (28%) MMUD, and 9 (9%) αβhaplo-HSCT patients died of transplant-related complications. With a median follow-up of 3.3 years, the 5-year probability of leukemia-free survival in the 3 groups was 67%, 55%, and 62%, respectively. In the 3 groups, chronic GVHD-free/relapse-free (GRFS) probability of survival was 61%, 34%, and 58%, respectively (P < .001). When compared with patients given MMUD-HSCT, αβhaplo-HSCT recipients had a lower cumulative incidence of nonrelapse mortality and a better GRFS (P < .001). These data indicate that αβhaplo-HSCT is a suitable therapeutic option for children with AL in need of transplantation, especially when an allele-matched UD is not available.
Collapse
|
9
|
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
|