1
|
Baranwal A, Graham C, Hassan K, Kassis R, Braun J, Bartoo G, Wolf R, He R, Viswanatha D, Matin A, Durani U, Kenderian S, Hefazi M, Mangaonkar AA, Shah MV, Litzow MR, Hogan WJ, Dingli D, Alkhateeb HB. Comparison of Chimerism Kinetics and Associated Outcomes in Patients receiving Post-Transplant Cyclophosphamide vs. Methotrexate based GVHD Prophylaxis following Allogeneic Hematopoietic Cell Transplant. Transplant Cell Ther 2025:S2666-6367(25)01167-4. [PMID: 40383196 DOI: 10.1016/j.jtct.2025.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 05/01/2025] [Accepted: 05/04/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION Post-transplant cyclophosphamide (PTCy) for graft-versus-host disease (GVHD) prophylaxis is now being used beyond haploidentical (HID) allogeneic hematopoietic cell transplant (alloHCT). However, the kinetics of chimerism in patients receiving PTCy and its impact on post-transplant relapse is unknown. In this study we describe the kinetics of donor chimerism in patients receiving PTCy, factors predisposing to mixed donor chimerism, and the associated survival outcomes. METHODS Patients undergoing alloHCT at Mayo Clinic, Rochester, from January 2018 to June 2023 were included in the study. Full donor chimerism was defined as donor cell fraction ≥95%, and mixed chimerism as donor cell fraction <95%. Analysis of covariance was used to assess the trend of tacrolimus levels in patients with mixed vs. full donor CD3 chimerism. Relapse-free survival (RFS) and overall survival (OS) from transplant were determined using the Kaplan-Meier method. Mixed donor chimerism was considered a time-dependent covariate in multivariate analysis. RESULTS A total of 500 patients were evaluated. A total of 189 (37.8%) patients received myeloablative conditioning (MAC); 27 (14.3%) of whom received PTCy and 162 (85.7%) received methotrexate (MTX) for GVHD prophylaxis. Among patients receiving PTCy, HID and mismatched donor transplants were significantly associated with a lower risk of mixed CD3 chimerism. In patients receiving PTCy, myeloablative busulfan/fludarabine (BuFlu), compared to non-busulfan MAC regimens, was associated with an increased risk of day +90 mixed chimerism (OR 10.47, P=0.02). However, reduced intensity (RIC) BuFlu was not associated with an increased risk of mixed CD3 chimerism (OR 0.71, P=0.7). Among patients receiving MAC and PTCy, those with high tacrolimus levels (≥11 mcg/ml) beyond the 2nd week post-transplant period were more likely to have mixed CD3 chimerism (F1,145 = 4.15, P=0.043). In patients receiving MAC and PTCy, day +90 mixed CD3 chimerism was associated with an inferior RFS (1-year RFS: 89.16% vs. 40.0%, P = 0.009). Multivariate analysis showed that mixed donor CD3 chimerism was associated with an inferior RFS in patients receiving MAC and PTCy (HR 6.53, 95% CI 1.18 - 36.15, P=0.032). CONCLUSION Among patients receiving MAC and PTCy, detection of mixed donor CD3 chimerism at any timepoint after transplant portends an inferior RFS. A high tacrolimus level beyond 2nd week of transplant in this subset of patients was associated with mixed CD3 chimerism. The detection of mixed CD3 chimerism provides an opportunity to implement strategies that may help in decreasing the risk of relapse in this subset of patients.
Collapse
Affiliation(s)
- Anmol Baranwal
- Division of Hematology, Mayo Clinic, Rochester, MN; Cancer Centers of Southwest Oklahoma, Lawton, OK
| | - Christopher Graham
- Division of Hematology, Mayo Clinic, Rochester, MN; Division of Hematology, Oncology and Transplantation, University of Minnesota, MN
| | | | - Rabee Kassis
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Jade Braun
- Department of Pharmacology, Mayo Clinic, Rochester, MN
| | | | - Robert Wolf
- Department of Pharmacology, Mayo Clinic, Rochester, MN
| | - Rong He
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, OK
| | | | - Aasyia Matin
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | | - David Dingli
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
2
|
Matsushima S, Kobayashi R, Sano H, Hori D, Yanagi M, Kobayashi K. Clinical Outcomes after One-day or Two-day Intervals in Conditioning Regimens for allo-HCT. J Pediatr Hematol Oncol 2023; 45:e378-e383. [PMID: 36161998 DOI: 10.1097/mph.0000000000002545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022]
Abstract
One-day or two-day intervals are generally inserted into scheduled conditioning regimens for allogeneic hematopoietic cell transplantation, primarily due to various social circumstances, such as unexpected natural adversities, abrupt deterioration of patient health, and delays in graft source arrival. We compared the clinical outcomes of patients with interrupted conditioning with those with ordinarily scheduled conditioning. We analyzed 83 patients (children and adolescents) with oncologic disease who underwent myeloablative conditioning with total body irradiation. Overall and event-free survival were similar between the groups ( P =0.955, P =0.908, respectively). Non-relapse mortality and relapse rates were similar between the groups ( P =0.923, P =0.946, respectively). The engraftment rate was not affected by interruption ( P =1.000). In contrast, the incidence of chronic graft-versus-host disease (GVHD) was higher in the interrupted group compared with the scheduled group, although there was no statistical significance (42% vs. 19%, P =0.063). Conditioning interruption was identified to be an independent risk factor for chronic GVHD by multivariate analysis (odds ratio: 3.72; 95% CI: 1.04 to 13.3; P =0.043). In conclusion, apart from the incidence of chronic GVHD, clinical outcomes were not affected by one-day or two-day intervals during conditioning.
Collapse
Affiliation(s)
- Satoru Matsushima
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
3
|
Klyuchnikov E, Badbaran A, Massoud R, Fritsche-Friedland U, Freiberger P, Ayuk F, Wolschke C, Bacher U, Kröger N. Post-transplant day +100 MRD detection rather than mixed chimerism predicts relapses after allo-SCT for intermediate risk AML patients transplanted in CR. Transplant Cell Ther 2022; 28:374.e1-374.e9. [DOI: 10.1016/j.jtct.2022.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 12/17/2022]
|
4
|
Kim J, Yun W, Park YJ, Seo J, Lee RDW, Shin S, Lee HJ, Kim IS, Choi JR, Lee ST. Chimerism Assay Using Single Nucleotide Polymorphisms Adjacent and in Linkage-Disequilibrium Enables Sensitive Disease Relapse Monitoring after Hematopoietic Stem-Cell Transplantation. Clin Chem 2021; 67:781-787. [PMID: 33582770 DOI: 10.1093/clinchem/hvab010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/31/2020] [Indexed: 11/14/2022]
Abstract
BACKGROUND Short tandem repeat (STR)-based chimerism analysis has been widely used for chimerism monitoring after hematopoietic stem-cell transplantation (HSCT), but technical artifacts can be problematic. We designed a chimerism assay using single nucleotide polymorphisms (SNPs) adjacent and in linkage-disequilibrium (CASAL), which doubly checked for SNP pairs, and thus could reduce background errors and increase analytical sensitivity. METHODS CASAL targeted 84 SNP pairs within 10 bp distance and in perfect linkage-disequilibrium. Using undiluted and serially diluted samples, baseline error rates, and linearity was calculated. Clinical performance of CASAL was evaluated in comparison with a conventional STR assay, using 191 posttransplant samples from 42 patients with HSCT. RESULTS CASAL had ∼10 times lower baseline error rates compared to that of ordinary next-generation sequencing. Limit of detection and quantification of CASAL were estimated to be 0.09 and 0.39%, respectively, with a linear range of 0.1-100%. CASAL correlated well with STR assay (r2 = 0.99) and the higher sensitivity enabled detection of low-level recipient chimerism and earlier prediction of relapse. CONCLUSIONS CASAL is a simple, analytically sensitive and accurate assay that can be used in clinical samples after HSCT with a higher performance compared to that of traditional assays. It should also be useful in other forensic and archeological testing.
Collapse
Affiliation(s)
- JinJu Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Woobin Yun
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
| | - Yu Jin Park
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Department of Laboratory Medicine, Armed Forces Yangju Hospital, Gyeonggi-do, South Korea
| | - Jieun Seo
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun-Ji Lee
- Department of Laboratory Medicine, Pusan National University, Yangsan Hospital, Yangsan, South Korea
| | - In Suk Kim
- Department of Laboratory Medicine, Pusan National University, School of Medicine, Pusan, South Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Dxome Co. Ltd, Seongnam-si, Gyeonggi-do, South Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Dxome Co. Ltd, Seongnam-si, Gyeonggi-do, South Korea
| |
Collapse
|
5
|
Imamura M. Impaired Hematopoiesis after Allogeneic Hematopoietic Stem Cell Transplantation: Its Pathogenesis and Potential Treatments. HEMATO 2021. [DOI: 10.3390/hemato2010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Impaired hematopoiesis is a serious complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Bone marrow aplasia and peripheral cytopenias arise from primary and secondary graft failure or primary and secondary poor graft function. Chimerism analysis is useful to discriminate these conditions. By determining the pathogenesis of impaired hematopoiesis, a timely and appropriate treatment can be performed. Hematopoietic system principally consists of hematopoietic stem cells and bone marrow microenvironment termed niches. Abnormality in hematopoietic stem and progenitor cells and/or abnormality in the relevant niches give rise to hematological diseases. Allo-HSCT is intended to cure each hematological disease, replacing abnormal hematopoietic stem cells and bone marrow niches with hematopoietic stem cells and bone marrow niches derived from normal donors. Therefore, treatment for graft failure and poor graft function after allo-HSCT is required to proceed based on determining the pathogenesis of impaired hematopoiesis. Recent progress in this area suggests promising treatment manipulations for graft failure and poor graft function.
Collapse
|
6
|
Gaut D, Mead M. Measurable residual disease in hematopoietic stem cell transplantation-eligible patients with acute myeloid leukemia: clinical significance and promising therapeutic strategies. Leuk Lymphoma 2020; 62:8-31. [DOI: 10.1080/10428194.2020.1827251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Daria Gaut
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Monica Mead
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
7
|
Mountjoy L, Palmer J, Kunze KL, Khera N, Sproat LZ, Leis JF, Noel P, Slack JL, Jain T. Does early chimerism testing predict outcomes after allogeneic hematopoietic stem cell transplantation? Leuk Lymphoma 2020; 62:252-254. [PMID: 33012186 DOI: 10.1080/10428194.2020.1827249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Luke Mountjoy
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Jeanne Palmer
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Katie L Kunze
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Phoenix, AZ, USA
| | - Nandita Khera
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Lisa Z Sproat
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Jose F Leis
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Pierre Noel
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - James L Slack
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Tania Jain
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
8
|
Fortschegger M, Preuner S, Printz D, Poetsch AR, Geyeregger R, Pichler H, Lawitschka A, Lion T. Detection and Monitoring of Lineage-Specific Chimerism by Digital Droplet PCR-Based Testing of Deletion/Insertion Polymorphisms. Biol Blood Marrow Transplant 2020; 26:1218-1224. [PMID: 32092354 DOI: 10.1016/j.bbmt.2020.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/03/2020] [Accepted: 02/13/2020] [Indexed: 12/31/2022]
Abstract
Analysis of specific leukocyte subsets for post-transplantation monitoring of chimerism provides greater sensitivity and clinical informativeness on dynamic changes in donor- and recipient-derived cells. Limitations of the most commonly used approach to chimerism testing relying on PCR-based analysis of microsatellite markers prompted us to assess the applicability of digital droplet (dd) PCR amplification of deletion/insertion polymorphisms (DIPs) for lineage-specific chimerism testing in the related stem cell transplantation setting, where the identification of informative markers facilitating the discrimination between donor-derived and recipient-derived cells can be challenging. We analyzed 100 genetically related patient-donor pairs by ddPCR analysis using commercially available DIP kits including large sets of polymorphic markers. At least 1 informative marker was identified in all related pairs analyzed, and 2 or more discriminating markers were detected in the majority (82%) of instances. The achievable detection limit is dependent on the number of cells available for analysis and was as low as 0.1% in the presence of ≥20,000 leukocytes available for DNA extraction. Moreover, the reproducibility and accuracy of quantitative chimerism analysis compared favorably to highly optimized microsatellite assays. Thus, the use of ddPCR-based analysis of DIP markers is an attractive approach to lineage-specific monitoring of chimerism in any allogeneic transplantation setting.
Collapse
Affiliation(s)
| | - Sandra Preuner
- St Anna Children's Cancer Research Institute, Vienna, Austria
| | - Dieter Printz
- St Anna Children's Cancer Research Institute, Vienna, Austria
| | - Anna R Poetsch
- St Anna Children's Cancer Research Institute, Vienna, Austria
| | - René Geyeregger
- St Anna Children's Cancer Research Institute, Vienna, Austria
| | | | | | - Thomas Lion
- St Anna Children's Cancer Research Institute, Vienna, Austria; Department of Pediatrics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Bouvier A, Riou J, Thépot S, Sutra Del Galy A, François S, Schmidt A, Orvain C, Estienne MH, Villate A, Luque Paz D, Cottin L, Ribourtout B, Beucher A, Delneste Y, Ifrah N, Ugo V, Hunault-Berger M, Blanchet O. Quantitative chimerism in CD3-negative mononuclear cells predicts prognosis in acute myeloid leukemia patients after hematopoietic stem cell transplantation. Leukemia 2019; 34:1342-1353. [PMID: 31768015 DOI: 10.1038/s41375-019-0624-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/18/2019] [Accepted: 11/03/2019] [Indexed: 12/20/2022]
Abstract
Relapse is a major complication of acute myeloid leukemia (AML) after allogeneic hematopoietic stem cell transplantation (SCT). The objective of our study was to evaluate chimerism monitoring on the CD3-negative mononuclear cells by RQ-PCR to predict relapse of patients allografted for AML and to compare its performance with WT1 quantification. A cohort of 100 patients undergoing allogenic SCT for AML was retrospectively analyzed in a single institution. Patients without complete chimerism, defined as less than 0.01% of recipient's DNA in CD3-negative cells, had a significantly higher risk of relapse and a lower overall survival (p < 0.001). An increase in the percentage of recipient DNA in CD3-negative cells was associated with an increased risk of relapse (p < 0.001) but not with overall survival. Comparable performances between monitoring of CD3-negative cell chimerism and WT1 expression to predict relapse was observed up to more than 90 days before hematological relapse, with sensitivity of 82% and 78%, respectively, and specificity of 100% for both approaches. Quantitative specific chimerism of the CD3-negative mononuclear fraction, enriched in blastic cells, is a new and powerful tool for monitoring measurable residual disease and could be used for AML patients without available molecular markers.
Collapse
Affiliation(s)
- Anne Bouvier
- CHU Angers, Laboratoire d'Hématologie, Angers, France. .,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France. .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | - Jérémie Riou
- MINT, UMR INSERM 1066, CNRS 6021, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Sylvain Thépot
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | | | - Sylvie François
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Aline Schmidt
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Corentin Orvain
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Marie-Hélène Estienne
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Tours, Service d'Hématologie biologique, Tours, France
| | - Alban Villate
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Tours, Service d'Hématologie et thérapie cellulaire, Tours, France
| | - Damien Luque Paz
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Laurane Cottin
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Bénédicte Ribourtout
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France
| | - Annaëlle Beucher
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France
| | - Yves Delneste
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,CHU Angers, Laboratoire d'Immunologie et Allergologie, Angers, France
| | - Norbert Ifrah
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Valérie Ugo
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Mathilde Hunault-Berger
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Odile Blanchet
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Centre de Ressources Biologiques, BB-0033-00038, Angers, France
| |
Collapse
|
10
|
Donor-Derived Cytokine-Induced Killer Cells after Nonmyeloablative Transplant for Myeloid Neoplasms. Biol Blood Marrow Transplant 2019; 25:e221-e222. [DOI: 10.1016/j.bbmt.2019.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/20/2019] [Indexed: 01/11/2023]
|
11
|
Comparison of reduced intensity conditioning regimens used in patients undergoing hematopoietic stem cell transplantation for myelofibrosis. Bone Marrow Transplant 2018; 54:204-211. [PMID: 29795431 DOI: 10.1038/s41409-018-0226-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023]
Abstract
The aim of this study is to compare clinical outcomes of patients who underwent allogeneic stem cell transplantation (HCT) for myelofibrosis with reduced intensity conditioning (RIC) using either Busulfan Fludarabine (BuFlu), Fludarabine Bis-chlorethyl-nitroso-urea/ carmustine Melphalan (FBM) or Fludarabine Melphalan (FluMel) regimens. Sixty-one patients were identified who underwent HCT with one of these RIC regimens. Overall survival (OS) was not different in the 3 groups. However, 100% donor chimerism was seen in more frequently at day +30 and day +100 in patients who received FBM or FluMel than BuFlu, in both CD3 and CD33 fractions. For instance, 100% donor chimerism in CD33 fraction was present in 100% patients in FBM cohort, 90% in FluMel cohort while 44% in BuFlu cohort at day +100. Acute graft-versus host disease, grade 2-4 and grade 3-4, was not statistically different in the 3 groups (BuFlu 47 and 35%, FBM 68 and 27%, FluMel 68 and 46%; p = 0.31 and 0.45). Relapses and non-relapse mortality was also not statistically significantly different. Our study shows similar OS with these 3 RIC regimens in myelofibrosis; although donor chimerism at day +30 and day +100 was better in patients who received FBM and FluMel.
Collapse
|
12
|
Bar M, Flowers MED, Storer BE, Chauncey TR, Pulsipher MA, Thakar MS, Bethge W, Storb R, Maloney DG, Sandmaier BM. Reversal of Low Donor Chimerism after Hematopoietic Cell Transplantation Using Pentostatin and Donor Lymphocyte Infusion: A Prospective Phase II Multicenter Trial. Biol Blood Marrow Transplant 2017; 24:308-313. [PMID: 29032276 DOI: 10.1016/j.bbmt.2017.10.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/09/2017] [Indexed: 11/17/2022]
Abstract
In a multicenter, prospective, phase II study we evaluated the safety and efficacy of pentostatin followed by donor lymphocyte infusion (DLI) in patients with low donor Tcell chimerism after allogeneic hematopoietic cell transplantation (HCT). Thirty-six patients with low donor blood CD3 chimerism were enrolled in this study. Thirty-five patients received a total of 41 DLIs after a dose of pentostatin, and 1 patient received pentostatin only. Median donor CD3 chimerism prompting the initiation of pentostatin and DLI was 28% (range, 5% to 47%). Responses (defined by increases in donor CD3 chimerism ≥10% maintained to day 56 post-DLI) were seen in 16 patients (44.4%) with a median rise in CD3 donor chimerism to 64% (range, 48% to 100%). There was a trend for better responses among 21 patients who received first treatment within 100 days after transplant (57% response rate) compared with15 patients who received first treatment more than 100 days after HCT (27% response rate, P = .07). Fourteen patients (39%) developed grades II to IV acute graft-versus-host disease (GVHD) at a median of 10 days (range, 0 to 83) after DLI. Ten patients (28%) developed extensive chronic GVHD. Seventeen patients (47%) developed new grade 4 cytopenias after DLI. There was no difference in relapse between nonresponders and responders. Twenty-eight patients (78%) died, most (n = 21) because of relapse. Five of 16 responders (31%) are alive, all disease-free, at a median of 60 months (range, 21 to 132) after DLI. Six of 20 nonresponders (30%) are alive at a median of 47 months (range, 16 to 100) after DLI, 3 in complete remission. Pentostatin and DLI had acceptable toxicity and appeared to increase low donor CD3 chimerism after HCT but had no impact on mortality.
Collapse
Affiliation(s)
- Merav Bar
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Mary E D Flowers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Barry E Storer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Thomas R Chauncey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington; VA Puget Sound Health Care System, Seattle, Washington
| | - Michael A Pulsipher
- Hematology, Oncology and Blood & Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, California
| | - Monica S Thakar
- Pediatric Blood & Marrow Transplant Program, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Wolfgang Bethge
- Medical Center, Department of Hematology and Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Rainer Storb
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - David G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Brenda M Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington.
| |
Collapse
|
13
|
Cytogenetic Evolution in Myeloid Neoplasms at Relapse after Allogeneic Hematopoietic Cell Transplantation: Association with Previous Chemotherapy and Effect on Survival. Biol Blood Marrow Transplant 2017; 23:782-789. [DOI: 10.1016/j.bbmt.2017.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/06/2017] [Indexed: 01/21/2023]
|
14
|
Gladstone DE, Petri M, Bolaños-Meade J, Dezern AE, Jones RJ, Fine D, Brodsky RA. Long-term systemic lupus erythematosus disease control after allogeneic bone marrow transplantation. Lupus 2016; 26:773-776. [PMID: 27687020 DOI: 10.1177/0961203316669242] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Systemic lupus erythematosus (SLE), a disorder of the immune system, is potentially curable by allogeneic bone marrow transplantation (alloBMT). Until recently, alloBMT was limited by donor availability and toxicity. Reduced intensity conditioning (RIC) combined with post-transplantation cyclophosphamide (PTCy) has improved the availability and safety of alloBMT permitting its exploration in severe-refractory autoimmune illnesses. We report the six-year follow-up of a young female whose refractory SLE-associated nephrosis resolved after RIC alloBMT with PTCy.
Collapse
Affiliation(s)
- D E Gladstone
- Johns Hopkins Hospital and Health System, Baltimore, MD, USA
| | - M Petri
- Johns Hopkins Hospital and Health System, Baltimore, MD, USA
| | - J Bolaños-Meade
- Johns Hopkins Hospital and Health System, Baltimore, MD, USA
| | - A E Dezern
- Johns Hopkins Hospital and Health System, Baltimore, MD, USA
| | - R J Jones
- Johns Hopkins Hospital and Health System, Baltimore, MD, USA
| | - D Fine
- Johns Hopkins Hospital and Health System, Baltimore, MD, USA
| | - R A Brodsky
- Johns Hopkins Hospital and Health System, Baltimore, MD, USA
| |
Collapse
|
15
|
Lee HC, Saliba RM, Rondon G, Chen J, Charafeddine Y, Medeiros LJ, Alatrash G, Andersson BS, Popat U, Kebriaei P, Ciurea S, Oran B, Shpall E, Champlin R. Mixed T Lymphocyte Chimerism after Allogeneic Hematopoietic Transplantation Is Predictive for Relapse of Acute Myeloid Leukemia and Myelodysplastic Syndromes. Biol Blood Marrow Transplant 2015; 21:1948-54. [PMID: 26183077 DOI: 10.1016/j.bbmt.2015.07.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/06/2015] [Indexed: 12/15/2022]
Abstract
Chimerism testing after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) represents a promising tool for predicting disease relapse, although its precise role in this setting remains unclear. We investigated the predictive value of T lymphocyte chimerism analysis at 90 to 120 days after allo-HSCT in 378 patients with AML/MDS who underwent busulfan/fludarabine-based myeloablative preparative regimens. Of 265 (70%) patients with available T lymphocyte chimerism data, 43% of patients in first or second complete remission (CR1/CR2) at the time of transplantation had complete (100%) donor T lymphocytes at day +90 to +120 compared with 60% of patients in the non-CR1/CR2 cohort (P = .005). In CR1/CR2 patients, donor T lymphocyte chimerism ≤ 85% at day +90 to +120 was associated with a higher frequency of 3-year disease progression (29%; 95% confidence interval [CI], 18% to 46% versus 15%; 95% CI, 9% to 23%; hazard ratio [HR], 2.1; P = .04). However, in the more advanced, non-CR1/CR2 cohort, mixed T lymphocyte chimerism was not associated with relapse (37%; 95% CI, 20% to 66% versus 34%; 95% CI, 25% to 47%; HR, 1.3; P = .60). These findings demonstrate that early T lymphocyte chimerism testing at day +90 to +120 is a useful approach for predicting AML/MDS disease recurrence in patients in CR1/CR2 at the time of transplantation.
Collapse
Affiliation(s)
- Hans C Lee
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rima M Saliba
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julianne Chen
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yasmeen Charafeddine
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gheath Alatrash
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan Ciurea
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Betul Oran
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
16
|
Bader P, Kreyenberg H, von Stackelberg A, Eckert C, Salzmann-Manrique E, Meisel R, Poetschger U, Stachel D, Schrappe M, Alten J, Schrauder A, Schulz A, Lang P, Müller I, Albert MH, Willasch AM, Klingebiel TE, Peters C. Monitoring of Minimal Residual Disease After Allogeneic Stem-Cell Transplantation in Relapsed Childhood Acute Lymphoblastic Leukemia Allows for the Identification of Impending Relapse: Results of the ALL-BFM-SCT 2003 Trial. J Clin Oncol 2015; 33:1275-84. [DOI: 10.1200/jco.2014.58.4631] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose To elucidate the impact of minimal residual disease (MRD) after allogeneic transplantation, the Acute Lymphoblastic Leukemia Berlin-Frankfurt-Münster Stem Cell Transplantation Group (ALL-BFM-SCT) conducted a prospective clinical trial. Patients and Methods In the ALL-BFM-SCT 2003 trial, MRD was assessed in the bone marrow at days +30, +60, +90, +180, and +365 after transplantation in 113 patients with relapsed disease. Standardized quantification of MRD was performed according to the guidelines of the Euro-MRD Group. Results All patients showed a 3-year probability of event-free survival (pEFS) of 55%. The cumulative incidence rates of relapse and treatment-related mortality were 32% and 12%, respectively. The pEFS was 60% for patients who received their transplantations in second complete remission, 50% for patients in ≥ third complete remission, and 0% for patients not in remission (P = .015). At all time points, the level of MRD was inversely correlated with event-free survival (EFS; P < .004) and positively correlated with the cumulative incidence of relapse (P < .01). A multivariable Cox model was fitted for each time point, which showed that MRD ≥ 10−4 leukemic cells was consistently correlated with inferior EFS (P < .003). The accuracy of MRD measurements in predicting relapse was investigated with time-dependent receiver operating curves at days +30, +60, +90, and +180. From day +60 onward, the discriminatory power of MRD detection to predict the probability of relapse after 1, 3, 6, and 9 months was more than 96%, more than 87%, more than 71%, and more than 61%, respectively. Conclusion MRD after transplantation was a reliable marker for predicting impending relapses and could thus serve as the basis for pre-emptive therapy.
Collapse
Affiliation(s)
- Peter Bader
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Hermann Kreyenberg
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Arend von Stackelberg
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Cornelia Eckert
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Emilia Salzmann-Manrique
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Roland Meisel
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Ulrike Poetschger
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Daniel Stachel
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Martin Schrappe
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Julia Alten
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Andre Schrauder
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Ansgar Schulz
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Peter Lang
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Ingo Müller
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Michael H. Albert
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Andre M. Willasch
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Thomas E. Klingebiel
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Christina Peters
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| |
Collapse
|
17
|
Frankfurt O, Zitzner JR, Tambur AR. Real-time qPCR for chimerism assessment in allogeneic hematopoietic stem cell transplants from unrelated adult and double umbilical cord blood. Hum Immunol 2015; 76:155-60. [DOI: 10.1016/j.humimm.2015.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 12/20/2014] [Accepted: 01/14/2015] [Indexed: 01/18/2023]
|
18
|
Martínez-Laperche C, Noriega V, Kwon M, Balsalobre P, González-Rivera M, Serrano D, Anguita J, Gayoso J, Díez-Martín JL, Buño I. Achievement of early complete donor chimerism in CD25+-activated leukocytes is a strong predictor of the development of graft-versus-host-disease after stem cell transplantation. Exp Hematol 2014; 43:4-13.e1. [PMID: 25450515 DOI: 10.1016/j.exphem.2014.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/05/2014] [Accepted: 09/06/2014] [Indexed: 12/22/2022]
Abstract
Chimerism dynamics in bone marrow, peripheral blood (PB), and T lymphocytes (TL) has been associated with the development of various complications after allogeneic stem-cell transplantation (allo-SCT). In the present study, the usefulness of chimerism monitoring in CD25(+)-activated leukocytes (AL), together with that in bone marrow, PB, and TL, for the anticipation of complications after allo-SCT, has been analyzed in 68 patients. In AL, we observed a slower dynamics toward complete chimerism (CC) than in PB (p = 0.042), while no significant differences were found between TL and PB (p = 0.12). Complete chimerism achievement in AL at day +30 has shown to be an independent risk factor for the development of grade II-IV acute graft-versus-host disease (aGvHD; hazard ratio [95% confidence interval]: 11.9 [1.5-91.7]; p = 0.017). Moreover, among patients achieving CC in TL and AL at different time-points after SCT (n = 17/68), the incidence of grade II-IV aGvHD was significantly higher in patients who achieved CC earlier in AL (5/5) than in those who achieved CC earlier in TL (1/11; p = 0.001). Therefore, achievement of early complete donor chimerism in CD25(+) AL is a strong predictor for the development of aGvHD. Prospective analysis of chimerism in AL could improve the post-SCT management of immunosuppressive therapy in transplanted patients.
Collapse
Affiliation(s)
- Carolina Martínez-Laperche
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Víctor Noriega
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Mi Kwon
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Pascual Balsalobre
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Milagros González-Rivera
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Sequencing and Genotyping Research Support Service, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - David Serrano
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Javier Anguita
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Jorge Gayoso
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - José Luis Díez-Martín
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ismael Buño
- Department of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
19
|
Zhao W, Wei L, Tan D, Su G, Zheng Y, He C, Mao ZJ, Singleton TP, Yin B. Cellular intrinsic mechanism affecting the outcome of AML treated with Ara-C in a syngeneic mouse model. PLoS One 2014; 9:e109198. [PMID: 25314317 PMCID: PMC4196759 DOI: 10.1371/journal.pone.0109198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 09/05/2014] [Indexed: 11/18/2022] Open
Abstract
The mechanisms underlying acute myeloid leukemia (AML) treatment failure are not clear. Here, we established a mouse model of AML by syngeneic transplantation of BXH-2 derived myeloid leukemic cells and developed an efficacious Ara-C-based regimen for treatment of these mice. We proved that leukemic cell load was correlated with survival. We also demonstrated that the susceptibility of leukemia cells to Ara-C could significantly affect the survival. To examine the molecular alterations in cells with different sensitivity, genome-wide expression of the leukemic cells was profiled, revealing that overall 366 and 212 genes became upregulated or downregulated, respectively, in the resistant cells. Many of these genes are involved in the regulation of cell cycle, cellular proliferation, and apoptosis. Some of them were further validated by quantitative PCR. Interestingly, the Ara-C resistant cells retained the sensitivity to ABT-737, an inhibitor of anti-apoptosis proteins, and treatment with ABT-737 prolonged the life span of mice engrafted with resistant cells. These results suggest that leukemic load and intrinsic cellular resistance can affect the outcome of AML treated with Ara-C. Incorporation of apoptosis inhibitors, such as ABT-737, into traditional cytotoxic regimens merits consideration for the treatment of AML in a subset of patients with resistance to Ara-C. This work provided direct in vivo evidence that leukemic load and intrinsic cellular resistance can affect the outcome of AML treated with Ara-C, suggesting that incorporation of apoptosis inhibitors into traditional cytotoxic regimens merits consideration for the treatment of AML in a subset of patients with resistance to Ara-C.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Biphenyl Compounds/pharmacology
- Cell Line, Tumor
- Cytarabine/pharmacology
- Cytarabine/therapeutic use
- Disease Models, Animal
- Down-Regulation/drug effects
- Drug Resistance, Neoplasm
- Gene Expression Profiling
- Inhibitor of Apoptosis Proteins/genetics
- Inhibitor of Apoptosis Proteins/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Nitrophenols/pharmacology
- Piperazines/pharmacology
- Sulfonamides/pharmacology
- Survival Rate
- Transplantation, Homologous
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Wenjun Zhao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Lirong Wei
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Dongming Tan
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Guangsong Su
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Yanwen Zheng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Chao He
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Zhengwei J. Mao
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center-Fairview, Minneapolis, Minnesota, United States of America
| | - Timothy P. Singleton
- Department of Laboratory of Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bin Yin
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, PR China
- Thrombosis and Hemostasis Key Lab of the Ministry of Health, Soochow University, Suzhou, Jiangsu Province, PR China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu Province, PR China
- * E-mail:
| |
Collapse
|
20
|
Adoptive therapy with donor lymphocyte infusion after allogenic hematopoietic SCT in pediatric patients. Bone Marrow Transplant 2014; 50:51-5. [PMID: 25310307 DOI: 10.1038/bmt.2014.200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 06/23/2014] [Accepted: 07/23/2014] [Indexed: 01/14/2023]
Abstract
The aim of this study was to analyse the experience of Polish Pediatric Group for Hematopoietic Stem Cell Transplantation in respect to donor lymphocyte infusion procedure. The study included 51 pediatric patients with malignant (45) and non-malignant (6) diseases treated with DLI in the period 1993-2012. The indications for DLI were as follows: (1) increasing recipient chimerism after non-ablative hematopoietic SCT (18 patients); (2) immunomodulation after a reduced intensity conditioning regimen (2 patients); (3) increase in minimal residual disease detection (3 patients); and (4) relapse (28 patients). DLI was carried out at a median of 6 (0.5-79) months after SCT. DLI was administered as either a single-dose (in 19 cases) or in escalating-dose regimens (in 32 cases). The median total dose of CD3-positive T cells was 28.0 (0.1-730.0) × 10(6)/kg body weight. The time for assessment of DLI efficacy ranged from 0 to 70 (median 3) months. At evaluation, 18 patients experienced CR, 3 achieved PR, 19 showed relapse and 11 rejected the graft. DLI was found to be effective in 39% of cases. Complications of the procedure occurred in 18 patients; of these, 2 died. To sum up DLI shows efficacy in a significant percentage of children. Mortality related to the therapy adverse effects is low. However, this method requires standardization.
Collapse
|
21
|
Reshef R, Hexner EO, Loren AW, Frey NV, Stadtmauer EA, Luger SM, Mangan JK, Gill SI, Vassilev P, Lafferty KA, Smith J, Van Deerlin VM, Mick R, Porter DL. Early donor chimerism levels predict relapse and survival after allogeneic stem cell transplantation with reduced-intensity conditioning. Biol Blood Marrow Transplant 2014; 20:1758-66. [PMID: 25016197 DOI: 10.1016/j.bbmt.2014.07.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/02/2014] [Indexed: 12/28/2022]
Abstract
The success of hematopoietic stem cell transplantation (HSCT) with reduced-intensity conditioning (RIC) is limited by a high rate of disease relapse. Early risk assessment could potentially improve outcomes by identifying appropriate patients for preemptive strategies that may ameliorate this high risk. Using a series of landmark analyses, we investigated the predictive value of early (day-30) donor chimerism measurements on disease relapse, graft-versus-host disease, and survival in a cohort of 121 patients allografted with a uniform RIC regimen. Chimerism levels were analyzed as continuous variables. In multivariate analysis, day-30 whole blood chimerism levels were significantly associated with relapse (hazard ratio [HR] = .90, P < .001), relapse-free survival (HR = .89, P < .001), and overall survival (HR = .94, P = .01). Day-30 T cell chimerism levels were also significantly associated with relapse (HR = .97, P = .002), relapse-free survival (HR = .97, P < .001), and overall survival (HR = .99, P = .05). Multivariate models that included T cell chimerism provided a better prediction for these outcomes compared with whole blood chimerism. Day-30 chimerism levels were not associated with acute or chronic graft-versus-host disease. We found that high donor chimerism levels were significantly associated with a low lymphocyte count in the recipient before transplant, highlighting the impact of pretransplant lymphopenia on the kinetics of engraftment after RIC HSCT. In summary, low donor chimerism levels are associated with relapse and mortality and can potentially be used as an early predictive and prognostic marker. These findings can be used to design novel approaches to prevent relapse and to improve survival after RIC HSCT.
Collapse
Affiliation(s)
- Ran Reshef
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Elizabeth O Hexner
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alison W Loren
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Noelle V Frey
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward A Stadtmauer
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Selina M Luger
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James K Mangan
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Saar I Gill
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Pavel Vassilev
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kathryn A Lafferty
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jacqueline Smith
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rosemarie Mick
- Department of Biostatistics & Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David L Porter
- Abramson Cancer Center and the Division of Hematology & Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
22
|
Comparison of chimerism and minimal residual disease monitoring for relapse prediction after allogeneic stem cell transplantation for adult acute lymphoblastic leukemia. Biol Blood Marrow Transplant 2014; 20:1522-9. [PMID: 24907626 DOI: 10.1016/j.bbmt.2014.05.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 05/27/2014] [Indexed: 12/17/2022]
Abstract
Little data are available on the relative merits of chimerism and minimal residual disease (MRD) monitoring for relapse prediction after allogeneic hematopoietic stem cell transplantation (HCT). We performed a retrospective analysis of serial chimerism assessments in 101 adult HCT recipients with acute lymphoblastic leukemia (ALL) and of serial MRD assessments in a subgroup of 22 patients. All patients had received myeloablative conditioning. The cumulative incidence of relapse was significantly higher in the patients with increasing mixed chimerism (in-MC) compared with those with complete chimerism, low-level MC, and decreasing MC, but the sensitivity of in-MC detection with regard to relapse prediction was only modest. In contrast, MRD assessment was highly sensitive and specific. Patients with MRD positivity after HCT had the highest incidence of relapse among all prognostic groups analyzed. The median time from MRD positivity to relapse was longer than the median time from detection of in-MC, but in some cases in-MC preceded MRD positivity. We conclude that MRD assessment is a powerful prognostic tool that should be included in the routine post-transplantation monitoring of patients with ALL, but chimerism analysis may provide additional information in some cases. Integration of these tools and clinical judgment should allow optimal decision making with regard to post-transplantation therapeutic interventions.
Collapse
|
23
|
Bernal T, Diez-Campelo M, Godoy V, Rojas S, Colado E, Alcoceba M, González M, Vidriales B, Sánchez-Guijo FM, López-Corral L, Luño E, del Cañizo C. Role of minimal residual disease and chimerism after reduced-intensity and myeloablative allo-transplantation in acute myeloid leukemia and high-risk myelodysplastic syndrome. Leuk Res 2014; 38:551-6. [DOI: 10.1016/j.leukres.2014.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 01/21/2014] [Accepted: 02/01/2014] [Indexed: 11/30/2022]
|
24
|
Chimerism status is correlated to acute graft-versus-host disease after allogeneic stem cell transplantation. Int J Hematol 2014; 99:323-8. [DOI: 10.1007/s12185-014-1510-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 01/09/2014] [Accepted: 01/15/2014] [Indexed: 10/25/2022]
|
25
|
Abstract
Molecular surveillance of hematopoietic chimerism is an important part of the routine diagnostic program in patients after allogeneic stem cell transplantation. Chimerism testing permits early prediction and documentation of successful engraftment and facilitates early risk assessment of impending graft rejection. In patients transplanted for treatment of malignant hematologic disorders, monitoring of chimerism can provide an early indication of incipient disease relapse. The investigation of chimerism has therefore become an indispensable tool for the management of patients during the post-transplant period. Increasing use of reduced-intensity conditioning, which is associated with prolonged duration of mixed hematopoietic chimerism, has further increased the clinical importance of chimerism analysis. At present, the most commonly used technical approach to the investigation of chimerism is microsatellite analysis by polymerase chain reaction. The investigation of chimerism within specific leukocyte subsets isolated from peripheral blood or bone marrow samples by flow sorting- or magnetic bead-based techniques provides more specific information on processes underlying the dynamics of donor/recipient chimerism. Moreover, cell subset-specific analysis permits the assessment of impending complications at a significantly higher sensitivity, thus providing a basis for earlier treatment decisions.
Collapse
Affiliation(s)
- Sandra Preuner
- Children's Cancer Research Institute (CCRI), Vienna, Austria
| | | |
Collapse
|
26
|
Hanson V, Adams B, Lord J, Barker A, Poulton K, Lee H. Assessment of the purity of isolated cell populations for lineage-specific chimerism monitoring post haematopoietic stem cell transplantation. ACTA ACUST UNITED AC 2013; 82:269-75. [DOI: 10.1111/tan.12172] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 05/31/2013] [Accepted: 07/02/2013] [Indexed: 11/30/2022]
Affiliation(s)
- V. Hanson
- Transplantation Laboratory, Manchester Royal Infirmary; Central Manchester University Hospitals NHS Trust; Manchester UK
| | - B. Adams
- Transplantation Laboratory, Manchester Royal Infirmary; Central Manchester University Hospitals NHS Trust; Manchester UK
| | - J. Lord
- Transplantation Laboratory, Manchester Royal Infirmary; Central Manchester University Hospitals NHS Trust; Manchester UK
| | - A. Barker
- Transplantation Laboratory, Manchester Royal Infirmary; Central Manchester University Hospitals NHS Trust; Manchester UK
| | - K. Poulton
- Transplantation Laboratory, Manchester Royal Infirmary; Central Manchester University Hospitals NHS Trust; Manchester UK
| | - H. Lee
- Transplantation Laboratory, Manchester Royal Infirmary; Central Manchester University Hospitals NHS Trust; Manchester UK
| |
Collapse
|
27
|
Thomson KJ, Peggs KS. Allogeneic transplantation in the UK: an aggregation of marginal gains? Br J Haematol 2013; 163:149-59. [PMID: 23889234 DOI: 10.1111/bjh.12497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A number of advances in clinical practice that are considered routine in modern allogeneic transplant programmes lack definitive supporting evidence, partly because they may offer modest incremental benefits that are difficult to demonstrate in a statistically robust manner given the relatively small cohorts of patients who undergo such procedures. Nevertheless, these marginal gains probably contribute therapeutically meaningful overall benefit, particularly when aggregated. We review the evidence for a number of these practices in terms of impact on transplant outcomes, with particular reference to the setting of T cell depletion as widely practiced in the United Kingdom, including high resolution tissue typing, surveillance for and therapy of infectious complications, chimerism-directed immune modulation and more sensitive monitoring for residual or progressive disease.
Collapse
Affiliation(s)
- Kirsty J Thomson
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | | |
Collapse
|
28
|
Combination of fludarabine, amsacrine, and cytarabine followed by reduced-intensity conditioning and allogeneic hematopoietic stem cell transplantation in patients with high-risk acute myeloid leukemia. Ann Hematol 2013; 92:1397-403. [DOI: 10.1007/s00277-013-1790-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 05/13/2013] [Indexed: 10/26/2022]
|
29
|
Rossi G, Carella AM, Minervini MM, Savino L, Fontana A, Pellegrini F, Greco MM, Merla E, Quarta G, Loseto G, Capalbo S, Palumbo G, Cascavilla N. Minimal residual disease after allogeneic stem cell transplant: a comparison among multiparametric flow cytometry, Wilms tumor 1 expression and chimerism status (Complete chimerism versus Low Level Mixed Chimerism) in acute leukemia. Leuk Lymphoma 2013; 54:2660-6. [PMID: 23547840 DOI: 10.3109/10428194.2013.789508] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Relapse represents the main cause of treatment failure after allogeneic stem cell transplant (allo-SCT). The detection of minimal residual disease (MRD) by multiparametric flow cytometry (MFC), chimerism, cytogenetics and molecular analysis may be critical to prevent relapse. Therefore, we assessed the overall agreement among chimerism (low level mixed chimerism [LL-MC] vs. complete chimerism [CC]), MFC and Wilms tumor 1 (WT1) mRNA to detect MRD and investigated the impact of MRD obtained from the three methods on patient outcome. Sixty-seven fresh bone marrow (BM) samples from 24 patients (17 acute myeloid leukemia [AML], seven acute lymphoblastic leukemia [ALL]) in complete remission (CR) after allo-SCT were investigated at different time points. A moderate agreement was found among the three techniques investigated. A higher concordance between positive results from MFC (75.0% vs. 32.7%, p = 0.010) and WT1 (58.3% vs. 29.1%, p = 0.090) was detected among LL-MC rather than CC samples. Relapse-free survival (RFS) and overall survival (OS) were found to be higher in MRD negative patients than in MRD positive patients analyzed with MFC and WT1. Our results discourage the use of low autologous signals as the only marker of MRD, and suggest the usefulness of MFC and WT1 real-time quantitative polymerase chain reaction (RQ-PCR) in stratifying patients with respect to risk of relapse.
Collapse
Affiliation(s)
- Giovanni Rossi
- Department of Hematology and Stem Cell Transplantation Unit
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Nikolousis E, Robinson S, Nagra S, Brookes C, Kinsella F, Tauro S, Jeffries S, Griffiths M, Mahendra P, Cook M, Paneesha S, Lovell R, Kishore B, Chaganti S, Malladi R, Raghavan M, Moss P, Milligan D, Craddock C. Post-transplant T cell chimerism predicts graft versus host disease but not disease relapse in patients undergoing an alemtuzumab based reduced intensity conditioned allogeneic transplant. Leuk Res 2013; 37:561-5. [PMID: 23395505 DOI: 10.1016/j.leukres.2013.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 01/12/2013] [Accepted: 01/13/2013] [Indexed: 11/18/2022]
Abstract
In this multicentre retrospective study we have studied the impact of T cell chimerism on the outcome of 133 patients undergoing an alemtuzumab based reduced intensity conditioning allograft (RIC). The median age of the patients was 50 years (range 42-55 years). 77 patients were transplanted using an HLA identical sibling donor while 56 patients received a fully matched volunteer unrelated donor graft. 64 patients had a lymphoid malignancy and 69 were transplanted for a myeloid malignancy. 38 patients (29%) relapsed with no significant difference in risk of relapse between patients developing full donor and mixed donor chimerism in the T-cell compartment on D+90 and D+180 post transplant. Day 90 full donor T cell chimerism correlated with an increased incidence of acute GVHD according to NIH criteria (p=0.0004) and the subsequent development of chronic GVHD. Consistent with previous observations, our results confirmed a correlation between the establishment of T cell full donor chimerism and acute GVHD in T deplete RIC allografts. However our study failed to identify any correlation between T cell chimerism and relapse risk and challenge the use of pre-emptive donor lymphocyte infusions (DLI) in patients with mixed T cell chimerism transplanted using an alemtuzumab based RIC regimen.
Collapse
Affiliation(s)
- E Nikolousis
- Department of Haematology, Heart of England NHS Foundation Trust, Birmingham, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Le Bourgeois A, Lestang E, Guillaume T, Delaunay J, Ayari S, Blin N, Clavert A, Tessoulin B, Dubruille V, Mahe B, Roland V, Gastinne T, Le Gouill S, Moreau P, Mohty M, Planche L, Chevallier P. Prognostic impact of immune status and hematopoietic recovery before and after fludarabine, IV busulfan, and antithymocyte globulins (FB2 regimen) reduced-intensity conditioning regimen (RIC) allogeneic stem cell transplantation (allo-SCT). Eur J Haematol 2013; 90:177-86. [DOI: 10.1111/ejh.12049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2012] [Indexed: 11/28/2022]
Affiliation(s)
- Amandine Le Bourgeois
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Elsa Lestang
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Thierry Guillaume
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Jacques Delaunay
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Sameh Ayari
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Nicolas Blin
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Aline Clavert
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Benoit Tessoulin
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Viviane Dubruille
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Beatrice Mahe
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Virginie Roland
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Thomas Gastinne
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Steven Le Gouill
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | - Philippe Moreau
- Centre Hospitalier et Universitaire (CHU) de Nantes, Hématologie Clinique; Centre d'Investigation Clinique en Cancérologie (CI2C); Nantes; France
| | | | - Lucie Planche
- Cellule de Promotion à la Recherche Clinique; CHU de Nantes; Nantes; France
| | | |
Collapse
|
32
|
Rauwerdink CA, Tsongalis GJ, Tosteson TD, Hill JM, Meehan KR. The practical application of chimerism analyses in allogeneic stem cell transplant recipients: Blood chimerism is equivalent to marrow chimerism. Exp Mol Pathol 2012; 93:339-44. [DOI: 10.1016/j.yexmp.2012.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 07/20/2012] [Indexed: 11/15/2022]
|
33
|
Machaczka M, Johansson JE, Remberger M, Hallböök H, Malm C, Lazarevic VL, Wahlin A, Omar H, Juliusson G, Kimby E, Hägglund H. Allogeneic hematopoietic stem cell transplant with reduced-intensity conditioning for chronic lymphocytic leukemia in Sweden: does donor T-cell engraftment 3 months after transplant predict survival? Leuk Lymphoma 2012; 53:1699-705. [DOI: 10.3109/10428194.2012.666661] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Salit RB, Fowler DH, Wilson WH, Dean RM, Pavletic SZ, Dunleavy K, Hakim F, Fry TJ, Steinberg SM, Hughes TE, Odom J, Bryant K, Gress RE, Bishop MR. Dose-adjusted EPOCH-rituximab combined with fludarabine provides an effective bridge to reduced-intensity allogeneic hematopoietic stem-cell transplantation in patients with lymphoid malignancies. J Clin Oncol 2012; 30:830-6. [PMID: 22312100 DOI: 10.1200/jco.2011.37.0296] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE There is currently no standard chemotherapy regimen for patients with lymphoid malignancies being considered for reduced-intensity conditioning allogeneic hematopoietic stem-cell transplantation (RIC-alloHSCT). The ideal regimen would provide disease control and result in lymphocyte depletion to facilitate engraftment. To this end, we developed a novel regimen by adding fludarabine to dose-adjusted continuous-infusion etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin plus with or without rituximab (DA-EPOCH-F/R). PATIENTS AND METHODS One hundred forty-seven patients with lymphoid malignancy (median age, 50 years) who had heavily pretreated (median prior regimens, three) and chemo-refractory (47%) disease were treated with DA-EPOCH-F/R before RIC-alloHSCT. Patients received one to three consecutive cycles until achieving lymphocyte depletion (CD4(+) count < 200/μL) or progressive disease. RESULTS Overall response rate was 41%; 39% of patients had stable disease. Toxicity included grade 4 neutropenia in 65% and thrombocytopenia in 25% of patients. DA-EPOCH-F/R resulted in lymphocyte depletion (P < .001), which was inversely associated with serum interleukin (IL) 7 and IL-15 levels. Of 147 patients, 143 patients proceeded to RIC-alloHSCT. Patients with lower CD3(+) (P < .001), CD4(+) (P < .001), and CD8(+) (P < .001) T-cell counts after DA-EPOCH-F/R were more likely to achieve full donor lymphoid chimerism by day +14 after transplant. Relative to nonresponders to DA-EPOCH-F/R, patients with complete and partial response had increased event-free survival (77.4 v 4.8 months; P < .001) and overall survival (98.5 v 16.2 months; P < .001). CONCLUSION DA-EPOCH-F/R safely provides tumor cytoreduction and lymphocyte depletion, thereby offering a bridge to RIC-alloHSCT in patients with aggressive lymphoid malignancies.
Collapse
Affiliation(s)
- Rachel B Salit
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Horky O, Mayer J, Kablaskova L, Razga F, Krejci M, Kissova J, Borsky M, Jeziskova I, Dvorakova D. Increasing hematopoietic microchimerism is a reliable indicator of incipient AML relapse. Int J Lab Hematol 2011; 33:57-66. [PMID: 20681999 DOI: 10.1111/j.1751-553x.2010.01249.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The reoccurrence or increase in autologous hematopoiesis after allogeneic transplantation has been linked to incipient leukemia relapse. However, the importance of such an emergency regarding microchimerism (i.e. mixed chimerism below 1% of autologous cells) still remains controversial, as fluctuating microchimerism can be observed for a very long time after transplantation. METHODS Using real-time PCR (RQ-PCR), we compare peripheral blood samples obtained from patients with acute myeloid leukemia (AML) before hematological relapse and those taken during complete remission (i.e. either complete cytogenetic remission or complete molecular remission where applicable). By comparison of these two groups, we describe microchimerism dynamics clearly connected with imminent AML relapse. Additionally, we compare applicability of RQ-PCR and conventional PCR with fragment analysis. RESULTS Mere reappearance of autologous hematopoiesis within patients with complete donor chimerism is alarming, and another sample with further increase confirms ongoing relapse. In case of patients with continuous microchimerism, another two consecutive samples with increasing trend are required. RQ-PCR predicted a significantly higher number of hematological relapses (87%vs. 39%) with a median anticipation period of 33 days, 26 days earlier than conventional PCR (P= 0.0002). Moreover, the outcome of microchimerism dynamics was in complete agreement with monitoring of minimal residual disease when analyzed from the same cell compartment. CONCLUSION Within this paper, we emphasize the importance of microchimerism monitoring as a reliable indicator of incipient AML relapse, especially in patients where no other specific molecular marker is available.
Collapse
Affiliation(s)
- O Horky
- Center of Molecular Biology and Gene Therapy, Department of Internal Medicine-Hematooncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Cernopolni 9, Brno, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Correlation Between the Kinetics of CD3+ Chimerism and the Incidence of Graft-Versus-Host Disease in Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplantation. Transplant Proc 2011; 43:1915-23. [DOI: 10.1016/j.transproceed.2011.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2010] [Revised: 01/20/2011] [Accepted: 02/02/2011] [Indexed: 11/20/2022]
|
37
|
Candoni A, Toffoletti E, Gallina R, Simeone E, Chiozzotto M, Volpetti S, Fanin R. Monitoring of minimal residual disease by quantitative WT1 gene expression following reduced intensity conditioning allogeneic stem cell transplantation in acute myeloid leukemia. Clin Transplant 2011; 25:308-16. [DOI: 10.1111/j.1399-0012.2010.01251.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Kröger N, Miyamura K, Bishop MR. Minimal residual disease following allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2011; 17:S94-100. [PMID: 21047560 PMCID: PMC3056549 DOI: 10.1016/j.bbmt.2010.10.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Minimal residual disease (MRD), both before and after transplantation, is a clinically important yet relatively poorly defined aspect of allogeneic hematopoietic stem cell transplantation (alloHSCT). The clinical relevance of MRD in the context of alloHSCT has been demonstrated by its association with the development of clinical relapse. However, with the possible exception of chronic myeloid leukemia (CML), the specific techniques, timing, frequency, and clinical utility, relative to improvement in patient outcomes, for monitoring MRD in the setting of alloHSCT has yet to be clearly defined. A concise overview of monitoring techniques for detecting MRD, as well as treatment strategies and biological and clinical research initiatives for MRD suggested by the National Cancer Institute First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation, is covered in this article.
Collapse
Affiliation(s)
| | | | - Michael R. Bishop
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
39
|
Kröger N, Bacher U, Bader P, Böttcher S, Borowitz MJ, Dreger P, Khouri I, Macapintac H, Olavarria E, Radich J, Stock W, Vose JM, Weisdorf D, Willasch A, Giralt S, Bishop MR, Wayne AS. NCI First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: report from the Committee on Disease-Specific Methods and Strategies for Monitoring Relapse following Allogeneic Stem Cell Transplantation. Part I: Methods, acute leukemias, and myelodysplastic syndromes. Biol Blood Marrow Transplant 2010; 16:1187-211. [PMID: 20558311 PMCID: PMC7272718 DOI: 10.1016/j.bbmt.2010.06.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 06/06/2010] [Indexed: 12/14/2022]
Abstract
Relapse has become the major cause of treatment failure after allogeneic stem cell transplantation. Outcome of patients with clinical relapse after transplantation generally remains poor, but intervention prior to florid relapse improves outcome for certain hematologic malignancies. To detect early relapse or minimal residual disease, sensitive methods such as molecular genetics, tumor-specific molecular primers, fluorescein in situ hybridization, and multiparameter flow cytometry (MFC) are commonly used after allogeneic stem cell transplantation to monitor patients, but not all of them are included in the commonly employed disease-specific response criteria. The highest sensitivity and specificity can be achieved by molecular monitoring of tumor- or patient-specific markers measured by polymerase chain reaction-based techniques, but not all diseases have such targets for monitoring. Similar high sensitivity can be achieved by determination of donor chimerism, but its specificity regarding detection of relapse is low and differs substantially among diseases. Here, we summarize the current knowledge about the utilization of such sensitive monitoring techniques based on tumor-specific markers and donor cell chimerism and how these methods might augment the standard definitions of posttransplant remission, persistence, progression, relapse, and the prediction of relapse. Critically important is the need for standardization of the different residual disease techniques and to assess the clinical relevance of minimal residual disease and chimerism surveillance in individual diseases, which in turn, must be followed by studies to assess the potential impact of specific interventional strategies.
Collapse
Affiliation(s)
- Nicolaus Kröger
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Germany
| | - Ulrike Bacher
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Germany
| | - Peter Bader
- Stem Cell Transplantation, Dept. of Pediatric Hematology/Oncology, University Hospital Frankfurt, Germany
| | - Sebastian Böttcher
- Second Department of Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | - Peter Dreger
- Department Medicine V, University of Heidelberg, Germany
| | - Issa Khouri
- Division of Hematology, M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Homer Macapintac
- Division of Nuclear Medicine, M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | - Jerald Radich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | | | - Julie M. Vose
- University of Nebraska Medical Center, Nebraska, Omaha, USA
| | | | - Andre Willasch
- Stem Cell Transplantation, Dept. of Pediatric Hematology/Oncology, University Hospital Frankfurt, Germany
| | - Sergio Giralt
- Division of Hematology, M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Michael R. Bishop
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, USA
| | - Alan S. Wayne
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, USA
| |
Collapse
|
40
|
Kern W, Bacher U, Haferlach C, Schnittger S, Haferlach T. The role of multiparameter flow cytometry for disease monitoring in AML. Best Pract Res Clin Haematol 2010; 23:379-90. [DOI: 10.1016/j.beha.2010.06.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
41
|
Jones CD, Arai S, Lowsky R, Tyan DB, Zehnder JL, Miklos DB. Complete donor T-cell engraftment 30 days after allogeneic transplantation predicts molecular remission in high-risk chronic lymphocytic leukaemia. Br J Haematol 2010; 150:637-9. [PMID: 20528878 PMCID: PMC2935248 DOI: 10.1111/j.1365-2141.2010.08252.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Carol D. Jones
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Sally Arai
- Department of Medicine, Division of Bone Marrow Transplantation, Stanford University School of Medicine, Stanford, CA
| | - Robert Lowsky
- Department of Medicine, Division of Bone Marrow Transplantation, Stanford University School of Medicine, Stanford, CA
| | - Dolly B. Tyan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - James L. Zehnder
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - David B. Miklos
- Department of Medicine, Division of Bone Marrow Transplantation, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
42
|
van Besien K, Dew A, Lin S, Joseph L, Godley LA, Larson RA, Odenike T, Rich E, Stock W, Wickrema A, Artz AS. Patterns and kinetics of T-cell chimerism after allo transplant with alemtuzumab-based conditioning: mixed chimerism protects from GVHD, but does not portend disease recurrence. Leuk Lymphoma 2010; 50:1809-17. [PMID: 19821799 DOI: 10.3109/10428190903200790] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We analyzed the kinetics of CD3 chimerism in 120 consecutive allogeneic hematopoietic cell transplantation (HCT) recipients receiving alemtuzumab-based conditioning. Fifty-two received fludarabine/melphalan, 44 received fludarabine/busulfan, and 24 received clofarabine/melphalan in addition to alemtuzumab. Post-transplant GVHD prophylaxis consisted of tacrolimus. No prophylactic donor lymphocyte infusion or other interventions were used for mixed donor chimerism (MDC). Bone marrow (BM) and/or peripheral blood (PB) samples were obtained at 30 days, 100 days, 180 days, and 1 year following HCT. On Day 30, 15% of assessable patients had MDC in the CD3 compartment. This had increased to 50% by Day 100, and to 63% by Day 180. MDC predicted for a lower risk of acute (p = 0.08) and particularly of chronic GVHD (p = 0.01). MDC was not associated with subsequent relapse or TRM (p = 0.67 and 0.72, respectively). A decline of more than 15% in CD3 chimerism between Day 30 and Day 180 predicted for a 40% risk of subsequent disease recurrence. The observation of MDC after alemtuzumab conditioning does not by itself constitute a risk factor for relapse and should not be used to guide therapeutic intervention. By contrast, declining donor chimerism between Day 30 and Day 180 is associated with a somewhat increased risk of disease recurrence. The high incidence of MDC after alemtuzumab containing conditioning contributes to the low risk of acute and chronic GVHD.
Collapse
Affiliation(s)
- Koen van Besien
- Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Because severe forms of the graft-versus-host reaction directed against normal tissues (also termed graft-versus-host disease [GVHD]) also contribute to morbidity and mortality following allogeneic hematopoietic stem cell transplantation, major efforts have focused on strategies to separate GVHD from the potentially beneficial immune reactivity against tumor (also called the graft-versus-tumor [GVT] effect). This article focuses on the data supporting the contribution of the GVT effect to cure of malignancy, what is known about the biology of the GVT reaction, and, finally, strategies to manipulate the GVT effect to increase the potency of HSCT.
Collapse
Affiliation(s)
- Terry J Fry
- Division of Blood and Marrow Transplantation/Immunology, Center for Cancer and Blood Disorders, Children's National Medical Center, 1 West Wing, 111 Michigan Avenue, NW, Washington, DC 20010, USA.
| | | | | |
Collapse
|
44
|
Villanueva FN, Pérez-Simón JA, Silva FF, Caballero-Velázquez TT, Sánchez-Guijo FF, Cañizo CC, Vázquez LL, Caballero DD, San Miguel JF. Oral Beclomethasone Dipropionate for the Treatment of Gastrointestinal Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2009; 15:1331-6. [DOI: 10.1016/j.bbmt.2009.05.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 05/26/2009] [Indexed: 11/16/2022]
|
45
|
Ringdén O, Karlsson H, Olsson R, Omazic B, Uhlin M. The allogeneic graft-versus-cancer effect. Br J Haematol 2009; 147:614-33. [PMID: 19735262 DOI: 10.1111/j.1365-2141.2009.07886.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Allogeneic haematological stem cell transplantation (HSCT) has developed into immunotherapy. Donor CD4+, CD8+ and natural killer (NK) cells have been reported to mediate graft-versus-leukaemia (GVL) effects, using Fas-dependent killing and perforin degranulation to eradicate malignant cells. Cytokines, such as interleukin-2, interferon-gamma and tumour necrosis factor-alpha potentiate the GVL effect. Post-transplant adoptive therapy of cytotoxic T-cells (CTL) against leukaemia-specific antigens, minor histocompatibility antigens, or T-cell receptor genes may constitute successful approaches to induce anti-tumour effects. Clinically, a significant GVL effect is induced by chronic rather than acute graft-versus-host disease (GVHD). An anti-tumour effect has also been reported for myeloma, lymphoma and solid tumours. Reduced intensity conditioning enables HSCT in older and disabled patients and relies on the graft-versus-tumour effect. Donor lymphocyte infusions promote the GVL effect and can be given as escalating doses with response monitored by minimal residual disease. A high CD34+ cell dose of peripheral blood stem cells increases GVL. There is a balance between effective immunosuppression, low incidence of GVHD and relapse. For instance, T-cell depletion of the graft increases the risk of relapse. This paper reviews the current knowledge in graft-versus-cancer effects. Future directions, such as immunotherapy using leukaemia-specific CTLs, allo-depleted T-cells and suicide gene manipulated T-cells, are presented.
Collapse
Affiliation(s)
- Olle Ringdén
- Centre for Allogeneic Stem Cell Transplantation and Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
46
|
Díez-Campelo M, Pérez-Simón JA, Ocio EM, Castilla C, González-Porras JR, Sánchez-Guijo FM, Vázquez L, Caballero MD, Cañizo MC, San Miguel JF. CD34?+ cell dose and outcome of patients undergoing reduced-intensity-conditioning allogeneic peripheral blood stem cell transplantation. Leuk Lymphoma 2009; 46:177-83. [PMID: 15621799 DOI: 10.1080/10428190400014900] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
While in bone marrow allogeneic transplantation the infusion of high doses of progenitor stem cells has a favourable impact on outcome, due to a faster hematopoietic and immune recovery, in the peripheral blood allo-setting the infusion of a high number of CD34 cells increases the risk of extensive chronic graft vs. host disease (cGVHD). This higher incidence of extensive cGVHD has an adverse impact on outcome due to a higher transplant related mortality, specially among patients receiving T-cell depleted allogeneic transplantation with myeloablative conditioning. By contrast, patients undergoing reduced intensity conditioning regimen may benefit from increasing higher CD34 + cell doses, especially those categorized as high risk according to disease status at transplant. Thus, the source of progenitors cells, type of conditioning and GVHD prophylaxis, among other factors, may influence the effect of the progenitor cell dose on outcome after allogeneic transplant.
Collapse
|
47
|
Díez-Campelo M, Pérez-Simón JA, Pérez J, Alcoceba M, Richtmon J, Vidriales B, San Miguel J. Minimal residual disease monitoring after allogeneic transplantation may help to individualize post-transplant therapeutic strategies in acute myeloid malignancies. Am J Hematol 2009; 84:149-52. [PMID: 19123459 DOI: 10.1002/ajh.21340] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This study evaluates the prognostic value of minimal residual disease (MRD) monitoring by multiparametric flow cytometry in 41 patients with acute myeloid leukemia or myelodysplastic syndrome undergoing allogeneic transplantation. MRD assessment after transplant (day +100) allowed to discriminate different risk populations, being the most significant cut-off value for outcome level of MRD < or > or = 10(-3). Outcome was significantly better among patients with low (<10(-3)) versus high (> or = 10(-3)) MRD at day +100 after transplant. Thus, overall survival was 73% versus 25% at 4 years among patients with low versus high MRD at day +100 after transplant (P = 0.002); 74% of patients with low MRD were event free at 4 years as compared to 17% among patients with high MRD (P = 0.01). In multivariate analysis, MRD value as well as chronic GVHD significantly influenced outcome. In conclusion, MRD monitoring early post-transplant is an important tool for outcome prediction and should be considered in decision making after allogeneic transplantation.
Collapse
Affiliation(s)
- María Díez-Campelo
- Servicio de Hematología, Hospital Universitario, CIC, IBMCC (USAL-CSIC), Salamanca, Spain
| | | | | | | | | | | | | |
Collapse
|
48
|
Lopez-Villar O, Garcia JL, Sanchez-Guijo FM, Robledo C, Villaron EM, Hernández-Campo P, Lopez-Holgado N, Diez-Campelo M, Barbado MV, Perez-Simon JA, Hernández-Rivas JM, San-Miguel JF, del Cañizo MC. Both expanded and uncultured mesenchymal stem cells from MDS patients are genomically abnormal, showing a specific genetic profile for the 5q− syndrome. Leukemia 2009; 23:664-72. [DOI: 10.1038/leu.2008.361] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
49
|
Minimal residual disease diagnostics in myeloid malignancies in the post transplant period. Bone Marrow Transplant 2008; 42:145-57. [PMID: 18587431 DOI: 10.1038/bmt.2008.185] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Allogeneic SCT is important in myelodysplastic syndrome, the BCR-ABL-negative chronic myeloproliferative diseases (CMPDs) and in poor-risk AML. Techniques to monitor the minimal residual disease, for example, by PCR or immunophenotyping gain increasing importance in the post transplantation period as basis for improved and earlier therapeutic interventions in impending relapse. Recent markers such as the NPM1 mutations in AML or the JAK2V617F mutation in the CMPD can be exactly quantified by real-time PCR and were evaluated for their prognostic value in the post transplantation phase and for their utility to plan adoptive immunotherapy in case of molecular relapse. With respect to chimerism, new and very sensitive methods were introduced, for example, quantitative assessment of genetic polymorphisms by real-time PCR, but also methods here are still highly individualized. Only in CML, where SCT focuses now on poor-risk cases or cases of tyrosine kinase inhibitor failure, follow-up schedules are standardized. Standardization of the different diagnostic techniques and of the intervals in the post transplantation period is urgently needed also in other myeloid malignancies and should be focus of future studies.
Collapse
|
50
|
A second prophylactic MHC-mismatched bone marrow transplantation protects against rat acute myeloid leukemia (BNML) without lethal graft-versus-host disease. Transplantation 2008; 85:102-11. [PMID: 18192919 DOI: 10.1097/01.tp.0000296856.53493.1f] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND We have employed a rat model for human acute myeloid leukemia, a promyelocytic leukemia in the BN rat strain (BNML), to develop new protocols for immunotherapy in combination with allogeneic bone marrow transplantation (alloBMT). The status of mixed chimerism in allotransplanted rats provided an opportunity for immunotherapy using alloreactive donor cells. In addition to T or natural killer (NK) cells, we introduced a second infusion of bone marrow cells as prophylactic donor lymphocyte infusions (DLI) to test whether an effective graft-versus-leukemia (GVL) response could be obtained without clinical graft-versus-host disease (GVHD). METHODS BN rats were sublethally irradiated and transplanted with T-cell depleted bone marrow cells from either fully major histocompatibility complex (MHC)-mismatched (PVG) donor rats or MHC-matched (PVG.1N) as controls. Seven days after transplantation, rats were given 500 leukemic cells to mimic minimal residual disease. Additional cellular therapy was given at day +7. The efficiency of DLI was monitored by chimerism analysis in peripheral blood. RESULTS Rats receiving infusions of NK cells succumbed to leukemia. T-DLI induced complete donor T-cell chimerism and lethal GVHD. A second alloBMT protected against leukemia. This effect was dependent on an MHC incompatibility between the donor and host and also on the presence of alloreactive T cells in the second bone marrow inoculum, resulting in an increased, mixed donor T-cell chimerism. CONCLUSION A second prophylactic transplantation influenced the degree of T-cell chimerism to balance favorably between GVL and GVHD. If applicable to humans, repeated alloBMT may provide a novel approach to leukemia therapy.
Collapse
|