1
|
Seferna K, Svaton M, Rennerova A, Skotnicova A, Reznickova L, Valova T, Sedlacek P, Riha P, Formankova R, Keslova P, Sramkova L, Stary J, Zuna J, Kolenova A, Salek C, Trka J, Fronkova E. NGS-MRD negativity in post-HSCT ALL spares unnecessary therapeutic interventions triggered by borderline qPCR results without an increase in relapse risk. Hemasphere 2025; 9:e70124. [PMID: 40201744 PMCID: PMC11978274 DOI: 10.1002/hem3.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/03/2025] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Monitoring of minimal residual disease (MRD) after hematopoietic stem cell transplantation (HSCT) in patients with acute lymphoblastic leukemia (ALL) is vital for timely therapeutic intervention planning. However, interpreting low-positive results from the current standard method, quantitative PCR (qPCR) of immunoglobulin and T-cell receptor gene rearrangements (IG/TR), poses challenges due to the risk of false positivity caused by non-specific amplification. We aimed to improve MRD detection specificity using the next-generation amplicon sequencing (NGS) of IG/TR rearrangements for better relapse prediction. In pediatric and young adult ALL patients undergoing sequential post-HSCT MRD monitoring, we prospectively re-tested positive non-quantifiable qPCR results with NGS-MRD using the EuroClonality-NGS approach. We were able to confirm 13 out of 47 (27.7%) qPCR positive results using the more specific NGS-MRD method. Out of 10 patients with at least one MRD positivity confirmed by NGS, six relapsed (60%) 1-3.7 months after testing. Among 25 patients with all NGS-MRD results negative, two relapses occurred (8%) after 5.1 and 12.1 months. One-year RFS was 40% versus 96% and 3-year OS was 33.3% versus 94.4% for the NGS-positive and NGS-negative groups, respectively. The difference was not attributable to a varying rate of therapeutic interventions. Six patients out of 14 who had immunosuppressive treatment tapered or received donor lymphocyte infusion in response to MRD positivity developed significant graft versus host disease, leading to one fatality. This underscores the importance of enhancing the post-HSCT relapse risk prediction accuracy through NGS-MRD testing to avoid unnecessary interventions.
Collapse
Affiliation(s)
- Krystof Seferna
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Michael Svaton
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Andrea Rennerova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Aneta Skotnicova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Leona Reznickova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Tatana Valova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petr Sedlacek
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petr Riha
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Renata Formankova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petra Keslova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Lucie Sramkova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Jan Stary
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Jan Zuna
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Alexandra Kolenova
- Department of Pediatric Hematology and OncologyNational Institute of Children's Diseases and Comenius UniversityBratislavaSlovakia
| | - Cyril Salek
- Institute of Haematology and Blood TransfusionPragueCzechia
- Institute of Clinical and Experimental HaematologyFirst Faculty of Medicine, Charles UniversityPragueCzechia
| | - Jan Trka
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Eva Fronkova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| |
Collapse
|
2
|
de Azambuja AP, Beltrame MP, Malvezzi M, Schluga YC, Justus JLP, Lima ACM, Funke VAM, Bonfim C, Pasquini R. Impact of high-sensitivity flow cytometry on peri-transplant minimal residual disease kinetics in acute leukemia. Sci Rep 2025; 15:6942. [PMID: 40011589 PMCID: PMC11865467 DOI: 10.1038/s41598-025-91936-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/24/2025] [Indexed: 02/28/2025] Open
Abstract
Minimal residual disease (MRD) detected before hematopoietic cell transplantation (HCT) is associated with adverse outcomes in patients with high-risk acute leukemia. However, the ideal time points for post-transplant MRD assessment and the clinical significance of low levels of residual disease in this context are unclear. We conducted a prospective real-world analysis of high-sensitivity flow cytometry MRD performed before and after transplant (at days 30, 60 and 100) in 77 acute leukemia patients. The aim was to evaluate the kinetics of disease elimination and correlate it with transplant outcomes. Pre-transplant MRD was negative in 42 (MRD-) and positive in 35 patients (MRD+). Post-transplant MRD assessment was feasible at day 30 (n = 30, 38.9%), day 60 (n = 27, 35.0%) and day 100 (n = 60, 77.9%). Relapses occurred in 8 patients in the MRD + group (22.9%) and three in the MRD-negative group (7.1%), p = 0.02. Pre-transplant MRD correlated with a decrease in overall survival (OS; 87.9% MRD- vs. 54.0% MRD+) and event-free survival (EFS; 85.3% MRD- vs. 51.1% MRD+), p = 0.001. Cumulative incidence of relapse (CIR) was 17.5% in MRD + vs. 2.6% in MRD- (p = 0.049). Non-relapse mortality (NRM) was 31.4% in MRD + vs. 12.1% in MRD- (p = 0.019). One-year OS was higher in patients with negative MRD at d100 (92.4%, 95% CI: 0.81-0.971) than positive d100 MRD (53.3%, 95% CI: 0.177-0.796), p < 0.0001. Disease status and d100 MRD were associated with OS, EFS and CIR. Differences in NRM between leukemia types (ALL: 18.9% MRD- vs. 50% MRD+, and AML 0% MRD- vs. 21.7% MRD+, p = 0.0158) were also observed. In conclusion, pre-transplant MRD assessed by highly sensitive flow cytometry accurately identified patients with adverse prognoses. Persistent MRD after HCT could predict relapse with high specificity and clinical sensitivity. These results highlight the importance of incorporating peri-transplant MRD kinetics into the routine treatment of acute leukemia, particularly in low/middle-income countries.
Collapse
Affiliation(s)
- Ana Paula de Azambuja
- Bone Marrow Transplantation Unit, Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil.
- Flow Cytometry Laboratory, Hospital de Clínicas Universidade Federal do Paraná, Avenida Nossa Senhora da Luz, 487, apto 601, 82510-020, Curitiba, Paraná, Brazil.
| | | | - Mariester Malvezzi
- Flow Cytometry Laboratory, Hospital de Clínicas Universidade Federal do Paraná, Avenida Nossa Senhora da Luz, 487, apto 601, 82510-020, Curitiba, Paraná, Brazil
| | - Yara Carolina Schluga
- Flow Cytometry Laboratory, Hospital de Clínicas Universidade Federal do Paraná, Avenida Nossa Senhora da Luz, 487, apto 601, 82510-020, Curitiba, Paraná, Brazil
| | - Julie Lillian Pimentel Justus
- Flow Cytometry Laboratory, Hospital de Clínicas Universidade Federal do Paraná, Avenida Nossa Senhora da Luz, 487, apto 601, 82510-020, Curitiba, Paraná, Brazil
| | | | | | - Carmem Bonfim
- Instituto de Pesquisa Pele Pequeno Príncipe/Faculdades Pequeno Principe Príncipe, Curitiba, Brazil
| | - Ricardo Pasquini
- Bone Marrow Transplantation Unit, Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil
| |
Collapse
|
3
|
Prockop S, Wachter F. The current landscape: Allogeneic hematopoietic stem cell transplant for acute lymphoblastic leukemia. Best Pract Res Clin Haematol 2023; 36:101485. [PMID: 37611999 DOI: 10.1016/j.beha.2023.101485] [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: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 08/25/2023]
Abstract
One of the consistent features in development of hematopoietic stem cell transplant (HCT) for Acute Lymphoblastic Leukemia (ALL) is the rapidity with which discoveries in the laboratory are translated into innovations in clinical care. Just a few years after murine studies demonstrated that rescue from radiation induced marrow failure is mediated by cellular not humoral factors, E. Donnall Thomas reported on the transfer of bone marrow cells into irradiated leukemia patients. This was followed quickly by the first descriptions of Graft versus Leukemia (GvL) effect and Graft versus Host Disease (GvHD). Despite the pivotal nature of these findings, early human transplants were uniformly unsuccessful and identified the challenges that continue to thwart transplanters today - leukemic relapse, regimen related toxicity, and GvHD. While originally only an option for young, fit patients with a matched family donor, expansion of the donor pool to include unrelated donors, umbilical cord blood units, and more recently the growing use of haploidentical donors have all made transplant a more accessible therapy for patients with ALL. Novel agents for conditioning, prevention and treatment of GvHD have improved outcomes and investigators continue to develop novel treatment strategies that balance regimen related toxicity with disease control. Our evolving understanding of how to prevent and treat GvHD and how to prevent relapse are incorporated into novel clinical trials that are expected to further improve outcomes. Here we review current considerations and future directions for both adult and pediatric patients undergoing HCT for ALL, including indication for transplant, donor selection, cytoreductive regimens, and outcomes.
Collapse
Affiliation(s)
- Susan Prockop
- Pediatric Stem Cell Transplant Program, DFCI/BCH Center for Cancer and Blood Disorders, Pediatrics, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States.
| | - Franziska Wachter
- Pediatric Stem Cell Transplant Program, DFCI/BCH Center for Cancer and Blood Disorders, Pediatrics, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States.
| |
Collapse
|
4
|
Rozmus J, Bhatt ST, Buxbaum NP, Cuvelier GDE, Li AM, Kitko CL, Schultz KR. Is It Possible to Separate the Graft-Versus-Leukemia (GVL) Effect Against B Cell Acute Lymphoblastic Leukemia From Graft-Versus-Host Disease (GVHD) After Hematopoietic Cell Transplant? Front Pediatr 2022; 10:796994. [PMID: 35402356 PMCID: PMC8987503 DOI: 10.3389/fped.2022.796994] [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: 10/18/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Hematopoietic cell transplant is a curative therapy for many pediatric patients with high risk acute lymphoblastic leukemia. Its therapeutic mechanism is primarily based on the generation of an alloreactive graft-versus-leukemia effect that can eliminate residual leukemia cells thus preventing relapse. However its efficacy is diminished by the concurrent emergence of harmful graft-versus-host disease disease which affects healthly tissue leading to significant morbidity and mortality. The purpose of this review is to describe the interventions that have been trialed in order to augment the beneficial graft-versus leukemia effect post-hematopoietic cell transplant while limiting the harmful consequences of graft-versus-host disease. This includes many emerging and promising strategies such as ex vivo and in vivo graft manipulation, targeted cell therapies, T-cell engagers and multiple pharmacologic interventions that stimulate specific donor effector cells.
Collapse
Affiliation(s)
- Jacob Rozmus
- Division of Hematology, Oncology and Bone Marrow Transplant, Department of Pediatrics, Faculty of Medicine, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Sima T Bhatt
- Washington University, Saint Louis, MO, United States
| | | | - Geoffrey D E Cuvelier
- Pediatric Blood and Marrow Transplantation, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Amanda M Li
- Division of Hematology, Oncology and Bone Marrow Transplant, Department of Pediatrics, Faculty of Medicine, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Carrie L Kitko
- Pediatric Hematology/Oncology Division, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kirk R Schultz
- Division of Hematology, Oncology and Bone Marrow Transplant, Department of Pediatrics, Faculty of Medicine, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
5
|
Cao LQ, Zhou Y, Liu YR, Xu LP, Zhang XH, Wang Y, Chen H, Chen YH, Wang FR, Han W, Sun YQ, Yan CH, Tang FF, Mo XD, Liu KY, Fan QZ, Chang YJ, Huang XJ. A risk score system for stratifying the risk of relapse in B cell acute lymphocytic leukemia patients after allogenic stem cell transplantation. Chin Med J (Engl) 2021; 134:1199-1208. [PMID: 33734137 PMCID: PMC8143760 DOI: 10.1097/cm9.0000000000001402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND For patients with B cell acute lymphocytic leukemia (B-ALL) who underwent allogeneic stem cell transplantation (allo-SCT), many variables have been demonstrated to be associated with leukemia relapse. In this study, we attempted to establish a risk score system to predict transplant outcomes more precisely in patients with B-ALL after allo-SCT. METHODS A total of 477 patients with B-ALL who underwent allo-SCT at Peking University People's Hospital from December 2010 to December 2015 were enrolled in this retrospective study. We aimed to evaluate the factors associated with transplant outcomes after allo-SCT, and establish a risk score to identify patients with different probabilities of relapse. The univariate and multivariate analyses were performed with the Cox proportional hazards model with time-dependent variables. RESULTS All patients achieved neutrophil engraftment, and 95.4% of patients achieved platelet engraftment. The 5-year cumulative incidence of relapse (CIR), overall survival (OS), leukemia-free survival (LFS), and non-relapse mortality were 20.7%, 70.4%, 65.6%, and 13.9%, respectively. Multivariate analysis showed that patients with positive post-transplantation minimal residual disease (MRD), transplanted beyond the first complete remission (≥CR2), and without chronic graft-versus-host disease (cGVHD) had higher CIR (P < 0.001, P = 0.004, and P < 0.001, respectively) and worse LFS (P < 0.001, P = 0.017, and P < 0.001, respectively), and OS (P < 0.001, P = 0.009, and P < 0.001, respectively) than patients without MRD after transplantation, transplanted in CR1, and with cGVHD. A risk score for predicting relapse was formulated with the three above variables. The 5-year relapse rates were 6.3%, 16.6%, 55.9%, and 81.8% for patients with scores of 0, 1, 2, and 3 (P < 0.001), respectively, while the 5-year LFS and OS values decreased with increasing risk score. CONCLUSION This new risk score system might stratify patients with different risks of relapse, which could guide treatment.
Collapse
Affiliation(s)
- Le-Qing Cao
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yang Zhou
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yan-Rong Liu
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Lan-Ping Xu
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yu Wang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Huan Chen
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yu-Hong Chen
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Feng-Rong Wang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Wei Han
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yu-Qian Sun
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Chen-Hua Yan
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Fei-Fei Tang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiao-Dong Mo
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Kai-Yan Liu
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Qiao-Zhen Fan
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, 2019RU029, Beijing, China
| |
Collapse
|
6
|
Merli P, Ifversen M, Truong TH, Marquart HV, Buechner J, Wölfl M, Bader P. Minimal Residual Disease Prior to and After Haematopoietic Stem Cell Transplantation in Children and Adolescents With Acute Lymphoblastic Leukaemia: What Level of Negativity Is Relevant? Front Pediatr 2021; 9:777108. [PMID: 34805054 PMCID: PMC8602790 DOI: 10.3389/fped.2021.777108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022] Open
Abstract
Minimal residual disease (MRD) assessment plays a central role in risk stratification and treatment guidance in paediatric patients with acute lymphoblastic leukaemia (ALL). As such, MRD prior to haematopoietic stem cell transplantation (HSCT) is a major factor that is independently correlated with outcome. High burden of MRD is negatively correlated with post-transplant survival, as both the risk of leukaemia recurrence and non-relapse mortality increase with greater levels of MRD. Despite growing evidence supporting these findings, controversies still exist. In particular, it is still not clear whether multiparameter flow cytometry and real-time quantitative polymerase chain reaction, which is used to recognise immunoglobulin and T-cell receptor gene rearrangements, can be employed interchangeably. Moreover, the higher sensitivity in MRD quantification offered by next-generation sequencing techniques may further refine the ability to stratify transplant-associated risks. While MRD quantification from bone marrow prior to HSCT remains the state of the art, heavily pre-treated patients may benefit from additional staging, such as using 18F-fluorodeoxyglucose positron emission tomography/computed tomography to detect focal residues of disease. Additionally, the timing of MRD detection (i.e., immediately before administration of the conditioning regimen or weeks before) is a matter of debate. Pre-transplant MRD negativity has previously been associated with superior outcomes; however, in the recent For Omitting Radiation Under Majority age (FORUM) study, pre-HSCT MRD positivity was associated with neither relapse risk nor survival. In this review, we discuss the level of MRD that may require pre-transplant therapy intensification, risking time delay and complications (as well as losing the window for HSCT if disease progression occurs), as opposed to an adapted post-transplant strategy to achieve long-term remission. Indeed, MRD monitoring may be a valuable tool to guide individualised treatment decisions, including tapering of immunosuppression, cellular therapies (such as donor lymphocyte infusions) or additional immunotherapy (such as bispecific T-cell engagers or chimeric antigen receptor T-cell therapy).
Collapse
Affiliation(s)
- Pietro Merli
- Department of Pediatric Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marianne Ifversen
- Pediatric Stem Cell Transplant and Immune Deficiency, Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tony H Truong
- Division of Pediatric Oncology and Bone Marrow Transplant, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| | - Hanne V Marquart
- Section for Diagnostic Immunology, Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Matthias Wölfl
- Pediatric Hematology, Oncology and Stem Cell Transplantation, Children's Hospital, Würzburg University Hospital, Würzburg, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, Goethe University, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
7
|
Wethmar K, Matern S, Eßeling E, Angenendt L, Pfeifer H, Brüggemann M, Stelmach P, Call S, Albring JC, Mikesch JH, Reicherts C, Groth C, Schliemann C, Berdel WE, Lenz G, Stelljes M. Monitoring minimal residual/relapsing disease after allogeneic haematopoietic stem cell transplantation in adult patients with acute lymphoblastic leukaemia. Bone Marrow Transplant 2020; 55:1410-1420. [PMID: 32001801 DOI: 10.1038/s41409-020-0801-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/14/2019] [Accepted: 01/16/2020] [Indexed: 12/22/2022]
Abstract
Relapse after allogeneic haematopoietic stem cell transplantation (SCT) is a major cause of death in patients with acute lymphoblastic leukaemia (ALL). Here, we retrospectively analysed the contributions of lineage-sorted donor cell chimerism (sDCC) and quantitative PCR (qPCR) targeting disease-specific genetic rearrangements to detect minimal residual/relapsing disease (MRD) and predict impending relapse in 94 adult ALL patients after SCT. With a median follow-up of surviving patients (n = 61) of 3.3 years, qPCR and/or sDCC measurements turned positive in 38 patients (40%). Of these, 22 patients relapsed and 16 remained in complete remission. At 3 years, qPCR and/or sDCC positive patients showed an increased incidence of relapse (50% vs. 4%, p < 0.0001), decreased relapse-free survival (RFS, 40% vs. 85%, p < 0.0001), and decreased overall survival (OS, 47% vs. 87%, p 0.004). Both, qPCR and sDCC pre-detected 11 of 21 relapses occurring within the first two years after SCT and, overall, complemented for each other method in four of the relapsing and four of the non-relapsing cases. Patients receiving pre-emptive MRD-driven interventions (n = 11) or not (n = 10) showed comparable median times until relapse, RFS, and OS. In our single centre cohort, qPCR and sDCC were similarly effective and complementary helpful to indicate haematological relapse of ALL after SCT.
Collapse
Affiliation(s)
- Klaus Wethmar
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Svenja Matern
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Eva Eßeling
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Linus Angenendt
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Heike Pfeifer
- Department of Haematology and Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Monika Brüggemann
- Department of Haematology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Patrick Stelmach
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Simon Call
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Jörn C Albring
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Jan-Henrik Mikesch
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Christian Reicherts
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Christoph Groth
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Christoph Schliemann
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Georg Lenz
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany
| | - Matthias Stelljes
- Department of Medicine A/Haematology and Oncology, University of Muenster, Muenster, Germany.
| |
Collapse
|
8
|
Cloos J, Ossenkoppele GJ, Dillon R. Minimal residual disease and stem cell transplantation outcomes. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:617-625. [PMID: 31808862 PMCID: PMC6913494 DOI: 10.1182/hematology.2019000006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Risk classification and tailoring of treatment are essential for improving outcome for patients with acute myeloid leukemia or high-risk myelodysplastic syndrome. Both patient and leukemia-specific characteristics assessed using morphology, cytogenetics, molecular biology, and multicolor flow cytometry are relevant at diagnosis and during induction, consolidation, and maintenance phases of the treatment. In particular, minimal residual disease (MRD) during therapy has potential as a prognostic factor of outcome, determination of response to therapy, and direction of targeted therapy. MRD can be determined by cell surface markers using multicolor flow cytometry, whereas leukemia-specific translocations and mutations are measured using polymerase chain reaction-based techniques and recently using next-generation sequencing. All these methods of MRD detection have their (dis)advantages, and all need to be standardized, prospectively validated, and improved to be used for uniform clinical decision making and a potential surrogate end point for clinical trials testing novel treatment strategies. Important issues to be solved are time point of MRD measurement and threshold for MRD positivity. MRD is used for stem cell transplantation (SCT) selection in the large subgroup of patients with an intermediate risk profile. Patients who are MRD positive will benefit from allo-SCT. However, MRD-negative patients have a better chance of survival after SCT. Therefore, it is debated whether MRD-positive patients should be extensively treated to become MRD negative before SCT. Either way, accurate monitoring of potential residual or upcoming disease is mandatory. Tailoring therapy according to MRD monitoring may be the most successful way to provide appropriate specifically targeted, personalized treatment.
Collapse
Affiliation(s)
- Jacqueline Cloos
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, VUMC, Amsterdam, The Netherlands; and
| | - Gert J Ossenkoppele
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, VUMC, Amsterdam, The Netherlands; and
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| |
Collapse
|
9
|
Lejman M, Zaucha-Prażmo A, Zawitkowska J, Mroczkowska A, Grabowski D, Kowalczyk JR, Drabko K. Impact of early chimerism status on clinical outcome in children with acute lymphoblastic leukaemia after haematopoietic stem cell transplantation. BMC Cancer 2019; 19:1141. [PMID: 31771553 PMCID: PMC6878687 DOI: 10.1186/s12885-019-6360-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 11/12/2019] [Indexed: 12/26/2022] Open
Abstract
Background The significance of very early chimerism assessment before day + 28, which is considered the moment of engraftment, is still unclear. In this retrospective study, we evaluated the clinical impact of very early chimerism on the clinical outcome after allogeneic haematopoietic stem cell transplantation (allo-HSCT) in children with acute lymphoblastic leukaemia (ALL). Methods The study group included 38 boys and 18 girls. Very early chimerism was evaluated on days + 7, + 14, + 21 and + 28 after the transplant. Short tandem repeat polymerase chain reaction (STR PCR) was used to analyse chimerism. Results Overall survival (OS) and event-free survival (EFS) were 84 and 80%, respectively. The OS in the group of 24 patients with complete donor chimerism on day + 14 was 83%, and it did not differ statistically compared to the 32 patients with mixed chimerism on day + 14 (OS was 84%). In our cohort of patients, the matched unrelated donor, male gender of donor, number of transplanted cells above 4.47 × 106 kg and no serotherapy with anti-thymocyte globulin (ATG) were statistically related to a higher level of donor chimerism. The immunophenotypes of disease, age of patient at time HSCT, recipient sex, stem cell source (peripheral blood/bone marrow) and conditioning regimen had no impact on early chimerism. Acute graft versus host disease grades II-IV was diagnosed in 23 patients who presented with donor chimerism levels above 60% on day 7. Conclusions The data presented in this study provide valuable insight into the analysis of very early chimerism in children with ALL treated with HSCT.
Collapse
Affiliation(s)
- Monika Lejman
- Laboratory of Genetic Diagnostics, Department of Pediatric Hematology, Oncology, and Transplantology, Medical University of Lublin, A. Gebali 6, 20-093, Lublin, Poland.
| | - Agnieszka Zaucha-Prażmo
- Department of Pediatric Hematology, Oncology, and Transplantology, Medical University of Lublin, A. Gebali 6, 20-093, Lublin, Poland
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology, and Transplantology, Medical University of Lublin, A. Gebali 6, 20-093, Lublin, Poland
| | - Aleksandra Mroczkowska
- Laboratory of Genetic Diagnostics, Department of Pediatric Hematology, Oncology, and Transplantology, Medical University of Lublin, A. Gebali 6, 20-093, Lublin, Poland
| | - Dominik Grabowski
- Department of Pediatric Hematology, Oncology, and Transplantology, Medical University of Lublin, A. Gebali 6, 20-093, Lublin, Poland
| | - Jerzy R Kowalczyk
- Department of Pediatric Hematology, Oncology, and Transplantology, Medical University of Lublin, A. Gebali 6, 20-093, Lublin, Poland
| | - Katarzyna Drabko
- Department of Pediatric Hematology, Oncology, and Transplantology, Medical University of Lublin, A. Gebali 6, 20-093, Lublin, Poland
| |
Collapse
|
10
|
Haugaard AK, Madsen HO, Marquart HV, Rosthøj S, Masmas TN, Heilmann C, Müller KG, Ifversen M. Highly sensitive chimerism detection in blood is associated with increased risk of relapse after allogeneic hematopoietic cell transplantation in childhood leukemia. Pediatr Transplant 2019; 23:e13549. [PMID: 31313439 DOI: 10.1111/petr.13549] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/04/2019] [Accepted: 06/19/2019] [Indexed: 12/19/2022]
Abstract
Analysis of chimerism in blood post-HCT using STR-PCR is routinely applied in parallel with quantification of MRD to predict relapse of leukemia. RQ-PCR chimerism is 10- to 100-fold more sensitive, but clinical studies in children are sparse. We analyzed IMC in blood samples following transplantation for acute lymphoblastic or myeloid leukemia in 56 children. IMC was defined as a minimum increase of (a) 0.1% or (b) 0.05% recipient DNA between two samples. The risk of relapse was higher in children with IMC of both 0.1% and 0.05% compared to children without IMC (HR 12.8 [95% CI: 3.9-41.4; P < .0001] and 7.6 [95% CI: 2.2-26.9; P < .01], respectively). The first IMC was detected at a median of 208 days prior to relapse. The 5-year cumulative incidence of relapse for children with a single IMC was 45.5% (CI 12.3-74.4) and 41.0% (14.2-66.6) for IMC above 0.1% and 0.05%, respectively. However, in 47 and 38 children never attaining IMC > 0.1% and >0.05%, 10 and 8 children relapsed, respectively. In a landmark analysis, no association was found between IMC prior to 90 days post-HCT and subsequent relapse by either classification of IMC and AUC for RQ-PCR chimerism was 54.2% (95 CI 27.7- 84.8). Although limited by a retrospective design, these results indicate that monitoring of RQ-PCR chimerism in peripheral blood may have a role in early detection of relapse in acute childhood leukemia.
Collapse
Affiliation(s)
- Anna Karen Haugaard
- Department for Children and Adolescents, Pediatric Hematopoietic Stem Cell Transplantation and Immunodeficiency, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hans Ole Madsen
- Department of Clinical Immunology, The Tissue Typing Laboratory, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hanne Vibeke Marquart
- Department of Clinical Immunology, The Tissue Typing Laboratory, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Susanne Rosthøj
- Department of Public Health, Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Tania Nicole Masmas
- Department for Children and Adolescents, Pediatric Hematopoietic Stem Cell Transplantation and Immunodeficiency, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Carsten Heilmann
- Department for Children and Adolescents, Pediatric Hematopoietic Stem Cell Transplantation and Immunodeficiency, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Klaus Gottlob Müller
- Department for Children and Adolescents, Pediatric Hematopoietic Stem Cell Transplantation and Immunodeficiency, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Marianne Ifversen
- Department for Children and Adolescents, Pediatric Hematopoietic Stem Cell Transplantation and Immunodeficiency, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
11
|
Cheng Y, Chen Y, Yan C, Wang Y, Zhao X, Chen Y, Han W, Xu L, Zhang X, Liu K, Wang S, Chang L, Xiao L, Huang X. Donor-Derived CD19-Targeted T Cell Infusion Eliminates B Cell Acute Lymphoblastic Leukemia Minimal Residual Disease with No Response to Donor Lymphocytes after Allogeneic Hematopoietic Stem Cell Transplantation. ENGINEERING 2019; 5:150-155. [DOI: 10.1016/j.eng.2018.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
12
|
Dubois V, Alizadeh M, Bourhis JH, Etancelin P, Farchi O, Ferrand C, Goursaud L, Mollet I, Renac V, Varlet P, Yakoub-Agha I, Bay JO. Étude du chimérisme après allogreffe de cellules hématopoïétiques : recommandations de la Société francophone de greffe de moelle et de thérapie cellulaire (SFGM-TC). Bull Cancer 2017; 104:S59-S64. [DOI: 10.1016/j.bulcan.2017.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/31/2017] [Accepted: 08/10/2017] [Indexed: 11/30/2022]
|
13
|
Minimal residual disease assessment by next-generation sequencing. Better tools to gaze into the crystal ball? Bone Marrow Transplant 2017; 52:952-954. [PMID: 28581473 DOI: 10.1038/bmt.2017.104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 11/08/2022]
|
14
|
Umeda K, Iwai A, Kawaguchi K, Mikami M, Nodomi S, Saida S, Hiramatsu H, Heike T, Ohmori K, Adachi S. Impact of post-transplant minimal residual disease on the clinical outcome of pediatric acute leukemia. Pediatr Transplant 2017; 21. [PMID: 28370903 DOI: 10.1111/petr.12926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/24/2017] [Indexed: 11/27/2022]
Abstract
This retrospective study examined the clinical significance of FCM-MRD in 36 patients with ALL and 29 patients with AML after their first allogeneic HSCT. Hematological (FCM-MRD ≥5.0%) and molecular relapse (FCM-MRD <5.0%) were first detected in 10 and two patients with ALL and in seven and eight patients with AML, respectively. Eight of 10 patients with molecular relapse eventually progressed to hematological relapse, although most were treated with immunological intervention by aggressive discontinuation of immunosuppressive therapy or donor lymphocyte infusion. Among these 12 patients, four of seven patients that obtained MRDneg CR following post-transplant chemotherapy remain alive and disease-free after their second HSCT; however, all five patients who underwent a second HSCT in non-CR died of disease or treatment-related complications. As the FCM-MRD monitoring system used in the current study was probably not sensitive enough to detect MRD, which could be elucidated by immunological intervention, more sensitive diagnostic tools are mandatory for post-transplant MRD monitoring. Additional studies are required to address the impact of presecond transplant MRD on the clinical outcome of second HSCT.
Collapse
Affiliation(s)
- Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Iwai
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Kawaguchi
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masamitsu Mikami
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seishiro Nodomi
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Saida
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidefumi Hiramatsu
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshio Heike
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsuyuki Ohmori
- Department of Clinical Laboratory, Kyoto University Hospital, Kyoto, Japan
| | - Souichi Adachi
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
15
|
Balduzzi A. The Value of Minimal Residual Disease (and Diamonds). Biol Blood Marrow Transplant 2017; 23:3-5. [DOI: 10.1016/j.bbmt.2016.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/11/2016] [Indexed: 11/25/2022]
|
16
|
Pre-Emptive Immunotherapy for Clearance of Molecular Disease in Childhood Acute Lymphoblastic Leukemia after Transplantation. Biol Blood Marrow Transplant 2016; 23:87-95. [PMID: 27742575 DOI: 10.1016/j.bbmt.2016.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/07/2016] [Indexed: 12/21/2022]
Abstract
Monitoring of minimal residual disease (MRD) or chimerism may help guide pre-emptive immunotherapy (IT) with a view to preventing relapse in childhood acute lymphoblastic leukemia (ALL) after transplantation. Patients with ALL who consecutively underwent transplantation in Frankfurt/Main, Germany between January 1, 2005 and July 1, 2014 were included in this retrospective study. Chimerism monitoring was performed in all, and MRD assessment was performed in 58 of 89 patients. IT was guided in 19 of 24 patients with mixed chimerism (MC) and MRD and by MRD only in another 4 patients with complete chimerism (CC). The 3-year probabilities of event-free survival (EFS) were .69 ± .06 for the cohort without IT and .69 ± .10 for IT patients. Incidences of relapse (CIR) and treatment-related mortality (CITRM) were equally distributed between both cohorts (without IT: 3-year CIR, .21 ± .05, 3-year CITRM, .10 ± .04; IT patients: 3-year CIR, .18 ± .09, 3-year CITRM .13 ± .07). Accordingly, 3-year EFS and 3-year CIR were similar in CC and MC patients with IT, whereas MC patients without IT experienced relapse. IT was neither associated with an enhanced immune recovery nor an increased risk for acute graft-versus-host disease. Relapse prevention by IT in patients at risk may lead to the same favorable outcome as found in CC and MRD-negative-patients. This underlines the importance of excellent MRD and chimerism monitoring after transplantation as the basis for IT to improve survival in childhood ALL.
Collapse
|
17
|
Pichler H, Fritsch G, König M, Daxberger H, Glogova E, Pötschger U, Breuer S, Lawitschka A, Güclü ED, Karlhuber S, Holter W, Haas OA, Lion T, Matthes-Martin S. Peripheral blood late mixed chimerism in leucocyte subpopulations following allogeneic stem cell transplantation for childhood malignancies: does it matter? Br J Haematol 2016; 173:905-17. [PMID: 26996395 DOI: 10.1111/bjh.14008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/03/2016] [Indexed: 01/26/2023]
Abstract
The impact of persistent mixed chimerism (MC) after haematopoietic stem cell transplantation (HSCT) remains unclarified. We investigated the incidence of MC in peripheral blood beyond day +50 after HSCT and its impact on rejection, chronic graft-versus-host disease (c-GvHD) and relapse in 161 children receiving allogeneic HSCT for haematological malignancies. The 1-year incidence of late MC was 26%. Spontaneous conversion to complete donor chimerism (CC) occurred in 43% of patients as compared to 62% after donor lymphocyte infusions. No graft rejection occurred. The 1-year incidence of c-GvHD was 20 ± 7% for MC, and 18 ± 4% for CC patients (P = 0·734). The 3-year cumulative incidence of relapse (CIR) according to chimerism status at days +50 and +100 was 22 ± 4% for CC patients vs. 22 ± 8% for MC patients (day +50; P = 0·935) and 21 ± 4% vs. 20 ± 7% (day +100; P = 0·907). Three-year CIRs in patients with persistent MC and patients with CC/limited MC were comparable (8 ± 7% vs. 19 ± 4%; P = 0·960). HSCT for acute leukaemia or myelodysplastic syndrome as secondary malignancies (hazard ratio (HR) 4·7; P = 0·008), for AML (HR 3·0; P = 0·02) and from mismatched donors (HR 3·1; P = 0·03) were independent factors associated with relapse. Our data suggest that late MC neither protects from c-GvHD nor does it reliably predict impending disease relapse.
Collapse
Affiliation(s)
- Herbert Pichler
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Gerhard Fritsch
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Margit König
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Helga Daxberger
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Evgenia Glogova
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Ulrike Pötschger
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Sabine Breuer
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Anita Lawitschka
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Ece D Güclü
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Susanne Karlhuber
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Holter
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Oskar A Haas
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria.,Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Thomas Lion
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Medical University of Vienna, Vienna, Austria
| | - Susanne Matthes-Martin
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Mellgren K, Arvidson J, Toporski J, Winiarski J. Chimerism analysis in clinical practice and its relevance for the detection of graft rejection and malignant relapse in pediatric hematopoietic stem cell transplant patients. Pediatr Transplant 2015; 19:758-66. [PMID: 26290161 DOI: 10.1111/petr.12580] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2015] [Indexed: 11/29/2022]
Abstract
Chimerism and clinical outcome data from 244 hematopoietic stem cell transplants in 218 children were retrospectively analyzed to assess their relevance for the detection of graft rejection and malignant relapse. Patients transplanted for a non-malignant disease had significantly higher proportions of residual recipient T cells in peripheral blood at one, three, and six months compared with patients transplanted for malignant disease. Recipient T-cell levels were below 50% at one month after transplantation in most patients (129 of 152 transplants). Graft rejection occurred more frequently in the group of patients with high levels of recipient cells at one month (10 graft rejections in the 23 patients with recipient T cells >50% at one month as compared to seven graft rejections occurred in 129 patients with recipient T cells <50% (p < 0.001). Multilineage chimerism data in 87 children with leukemia at one, three, and six months after transplantation were not correlated with subsequent relapse of malignant disease. In conclusion, early analysis of lineage-specific chimerism in peripheral blood can be used to identify patients who are at high risk of graft rejection. However, the efficacy of early chimerism analysis for predicting leukemia relapse was limited.
Collapse
Affiliation(s)
- Karin Mellgren
- Department of Pediatrics, Institution for Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Johan Arvidson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Jacek Toporski
- Section of Pediatric Oncology/Hematology, Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - Jacek Winiarski
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|