1
|
Puzo CJ, Siddon AJ. Chimerism testing in myeloid malignancies: techniques, considerations, and connections to post-transplant outcomes. Pathology 2025; 57:267-275. [PMID: 39934013 DOI: 10.1016/j.pathol.2024.12.632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 02/13/2025]
Abstract
Relapse represents the most significant cause of death post-allogeneic stem cell transplant (aSCT) for patients with myeloid malignancies. Early intervention among patients deemed high risk for relapse is one focus to improve aSCT-related outcomes. As such, the early identification of residual malignant cell burden is of critical importance. Chimerism testing, the assessment of the relative percentage of donor to recipient haematopoietic cells based on genetic differences, offers one practical means to assess for residual host haematopoietic cells, which could be indicative of leukaemic cell burden. The purpose of this review paper is to discuss the use of chimerism testing to better understand the risk of relapse. Important consideration will be given to the various laboratory techniques used to compute donor chimerism percentage and their relative limitations, the external factors that may influence the relative value of donor to recipient chimerism and its importance among the major myeloid malignancy diagnostic categories. Special attention will be paid to strategies to improve the sensitivity and specificity of chimerism testing, namely its concurrent use with measurable residual disease and the use of lineage-specific chimerism to both improve relapse detection and guide therapy decisions.
Collapse
Affiliation(s)
| | - Alexa J Siddon
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Delie A, Verlinden A, Beel K, Deeren D, Mazure D, Baron F, Breems D, De Becker A, Graux C, Lewalle P, Maertens J, Poire X, Schoemans H, Selleslag D, Van Obbergh F, Kerre T. Use of chimerism analysis after allogeneic stem cell transplantation: Belgian guidelines and review of the current literature. Acta Clin Belg 2021; 76:500-508. [PMID: 32362204 DOI: 10.1080/17843286.2020.1754635] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment option in both adult and pediatric patients with malignant and non-malignant hematological diseases. Chimerism analysis, which determines the donor or recipient origin of hematopoietic cells in HSCT recipients, is an essential aspect of post-HSCT follow-up.Objectives: To review the current literature and develop Belgian consensus guidelines for the use of chimerism analysis in the standard of care after allogeneic HSCT.Methods: Non-systematic review of the literature in consultancy with the members of the BHS transplantation committee.Results: Clinical application with regards to prediction of graft failure or relapse as well as cell source are reviewed. A consensus guideline on the use of chimerism analysis after HSCT is presented.Conclusion: Monitoring of the dynamics or kinetics of a patient's chimerism status by serial analysis at fixed time points, as well as on suspicion of relapse or graft failure, is needed to monitor engraftment levels, as well as disease control and possible relapse.
Collapse
Affiliation(s)
- Anke Delie
- Department of Hematology, University Hospital, Ghent University, Ghent, Belgium
| | - Anke Verlinden
- Department of Hematology, University Hospital, University of Antwerp, Antwerp, Belgium
| | - Karolien Beel
- Department of Hematology, Ziekenhuis Netwerk, Antwerpen, Belgium
| | - Dries Deeren
- Department of Hematology, AZ Delta, Roeselare, Belgium
| | - Dominiek Mazure
- Department of Hematology, University Hospital, Ghent University, Ghent, Belgium
| | - Frédéric Baron
- Department of Hematology, University Hospital, University of Liège, Liège, Belgium
| | - Dimitri Breems
- Department of Hematology, Ziekenhuis Netwerk, Antwerpen, Belgium
| | - Ann De Becker
- Department of Hematology, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carlos Graux
- Department of Hematology, University Hospital Namur, Université Catholique de Louvain, Belgium
| | - Philippe Lewalle
- Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Johan Maertens
- Department of Hematology, University Hospital, KU Leuven, Leuven, Belgium
| | - Xavier Poire
- Department of Hematology, University Hospital Saint Luc, Univeristé Catholique de Louvain, Brussels, Belgium
| | - Helene Schoemans
- Department of Hematology, University Hospital, KU Leuven, Leuven, Belgium
| | | | | | - Tessa Kerre
- Department of Hematology, University Hospital, Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
Clonal expansion of CD8+ T cells reflects graft-versus-leukemia activity and precedes durable remission following DLI. Blood Adv 2021; 5:4485-4499. [PMID: 34535011 PMCID: PMC8579265 DOI: 10.1182/bloodadvances.2020004073] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/05/2021] [Indexed: 11/30/2022] Open
Abstract
GVL activity in patients receiving DLI for relapse after allogeneic hematopoietic stem cell transplantation is associated with clonal expansion of CD8+ T cells. Absence of clonal expansion of the CD8+TRB repertoire after DLI predicts relapse at a median time of 11.2 months before clinical diagnosis.
Donor lymphocyte infusion (DLI) is a standard of care for relapse of acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. Currently it is poorly understood how and when CD8+ αβ T cells exert graft-versus-leukemia (GVL) activity after DLI. Also, there is no reliable biomarker to monitor GVL activity of the infused CD8+ T cells. Therefore, we analyzed the dynamics of CD8+ αβ T-cell clones in patients with DLI. In this prospective clinical study of 29 patients, we performed deep T-cell receptor β (TRB ) sequencing of sorted CD8+ αβ T cells to track patients’ repertoire changes in response to DLI. Upon first occurrence of GVL, longitudinal analyses revealed a preferential expansion of distinct CD8+TRB clones (n = 14). This did not occur in samples of patients without signs of GVL (n = 11). Importantly, early repertoire changes 15 days after DLI predicted durable remission for the 36-month study follow-up. Furthermore, absence of clonal outgrowth of the CD8+TRB repertoire after DLI was an early biomarker that predicted relapse at a median time of 11.2 months ahead of actual diagnosis. Additionally, unbiased sample analysis regardless of the clinical outcome revealed that patients with decreasing CD8+TRB diversity at day 15 after DLI (n = 13) had a lower relapse incidence (P = .0040) compared with patients without clonal expansion (n = 6). In conclusion, CD8+TRB analysis may provide a reliable tool for predicting the efficacy of DLI and holds the potential to identify patients at risk for progression and relapse after DLI.
Collapse
|
4
|
Tyler J, Kumer L, Fisher C, Casey H, Shike H. Personalized Chimerism Test that Uses Selection of Short Tandem Repeat or Quantitative PCR Depending on Patient's Chimerism Status. J Mol Diagn 2019; 21:483-490. [PMID: 30797064 DOI: 10.1016/j.jmoldx.2019.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 01/14/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
Chimerism testing is used to monitor engraftment and risk of relapse after allogeneic hematopoietic stem cell transplantation for hematologic malignancies. Although short tandem repeat (STR) method is widely used among clinical laboratories, quantitative PCR (qPCR) provides better sensitivity (0.1%) than STR (1% to 5%) but is less accurate than STR for patients in mixed chimerism. qPCR chimerism allows evaluation of residual recipient cells as a surrogate of measurable residual disease. To achieve higher sensitivity and accuracy, we applied qPCR or STR based on patient chimerism status (recipient alleles <5% or ≥5%, respectively). Of the 230 patients tested by STR in a 1-year period, excluding 10 deceased patients, 30 qPCR markers were genotyped and 167 patients converted to qPCR chimerism (76%), including eight patients undergoing multiple-donor transplantation. STR was continued on 53 patients (24%) for the following reasons: mixed chimerism (n = 23), lack of donor or pretransplantation DNA (n = 22), and insufficient qPCR informative markers [8 of 60 patients with related donors (13.3%)]. qPCR detected residual recipient chimerism in 85.5% of patients with complete chimerism by STR (<5% recipient). Selecting STR or qPCR testing based on each patient's chimerism status facilitates sensitive and accurate chimerism testing in clinical settings. In addition, we discuss clinical relevance of chimerism testing for measurable residual disease detection in various hematologic malignancies.
Collapse
Affiliation(s)
- Jennifer Tyler
- Department of Pathology, Histocompatibility, and Immunogenetics, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Lorie Kumer
- Department of Pathology, Histocompatibility, and Immunogenetics, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Carolyn Fisher
- Department of Pathology, Histocompatibility, and Immunogenetics, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Heather Casey
- Department of Pathology, Histocompatibility, and Immunogenetics, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Hiroko Shike
- Department of Pathology, Histocompatibility, and Immunogenetics, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania.
| |
Collapse
|
5
|
Rautenberg C, Germing U, Haas R, Kobbe G, Schroeder T. Relapse of Acute Myeloid Leukemia after Allogeneic Stem Cell Transplantation: Prevention, Detection, and Treatment. Int J Mol Sci 2019; 20:E228. [PMID: 30626126 PMCID: PMC6337734 DOI: 10.3390/ijms20010228] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a phenotypically and prognostically heterogeneous hematopoietic stem cell disease that may be cured in eligible patients with intensive chemotherapy and/or allogeneic stem cell transplantation (allo-SCT). Tremendous advances in sequencing technologies have revealed a large amount of molecular information which has markedly improved our understanding of the underlying pathophysiology and enables a better classification and risk estimation. Furthermore, with the approval of the FMS-like tyrosine kinase 3 (FLT3) inhibitor Midostaurin a first targeted therapy has been introduced into the first-line therapy of younger patients with FLT3-mutated AML and several other small molecules targeting molecular alterations such as isocitrate dehydrogenase (IDH) mutations or the anti-apoptotic b-cell lymphoma 2 (BCL-2) protein are currently under investigation. Despite these advances, many patients will have to undergo allo-SCT during the course of disease and depending on disease and risk status up to half of them will finally relapse after transplant. Here we review the current knowledge about the molecular landscape of AML and how this can be employed to prevent, detect and treat relapse of AML after allo-SCT.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Hematopoietic Stem Cell Transplantation
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/prevention & control
- Leukemia, Myeloid, Acute/therapy
- Mutation/genetics
- Neoplasm, Residual/diagnosis
- Neoplasm, Residual/drug therapy
- Recurrence
- Transplantation, Homologous
Collapse
Affiliation(s)
- Christina Rautenberg
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| |
Collapse
|
6
|
Diagnostic value of highly-sensitive chimerism analysis after allogeneic stem cell transplantation. Bone Marrow Transplant 2018; 53:1457-1465. [DOI: 10.1038/s41409-018-0176-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 03/11/2018] [Accepted: 03/17/2018] [Indexed: 12/18/2022]
|
7
|
Chimerism in Myeloid Malignancies following Stem Cell Transplantation Using FluBu4 with and without Busulfan Pharmacokinetics versus BuCy. Adv Hematol 2017; 2017:8690416. [PMID: 29250116 PMCID: PMC5698787 DOI: 10.1155/2017/8690416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/25/2017] [Accepted: 10/10/2017] [Indexed: 11/17/2022] Open
Abstract
In the era of precision medicine, the impact of personalized dosing of busulfan is not clear. We undertook a retrospective analysis of 78 patients with myeloid malignancies who received fludarabine and busulfan (FluBu4) with or without measuring Bu pharmacokinetics (Bu PK) and those who received busulfan with cyclophosphamide (BuCy). Fifty-five patients received FluBu4, of whom 21 had Bu PK measured, and 23 patients received BuCy. Total donor cell chimerism showed that the percentage of patients maintaining 100% donor chimerism on day 100 was 66.7%, 38.2%, and 73.9% in the FluBu4 with PK, FluBu4 with no PK, and BuCy, respectively (P = .001). Patients who had decreasing donor chimerism by day 100 were 23.8%, 52.9%, and 26.1% in the FluBu4 with PK, FluBu4 with no PK, and BuCy, respectively (P = .04). Bu PK group had fewer patients with less than 95% donor chimerism on day 30, which was not statistically significant, 5% (FluBu4 PK), 31% (FluBu4 with no PK), and 21% (BuCy) (P = .18). Survival distributions were not statistically significant (P = .11). Thus, personalized drug dosing can impact donor chimerism in myeloid malignancies. This will need to be examined in larger retrospective multicenter studies and prospective clinical trials.
Collapse
|
8
|
Third International workshop on the biology, prevention, and treatment of relapse after stem cell transplantation. Bone Marrow Transplant 2017; 53:1-2. [DOI: 10.1038/bmt.2017.218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
9
|
Mo XD, Lv M, Huang XJ. Preventing relapse after haematopoietic stem cell transplantation for acute leukaemia: the role of post-transplantation minimal residual disease (MRD) monitoring and MRD-directed intervention. Br J Haematol 2017; 179:184-197. [PMID: 28542711 DOI: 10.1111/bjh.14778] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Xiao-Dong Mo
- Peking University People's Hospital; Peking University Institute of Haematology; Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation; Beijing China
| | - Meng Lv
- Peking University People's Hospital; Peking University Institute of Haematology; Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation; Beijing China
| | - Xiao-Jun Huang
- Peking University People's Hospital; Peking University Institute of Haematology; Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation; Beijing China
- Peking-Tsinghua Centre for Life Sciences; Beijing China
| |
Collapse
|
10
|
van der Torren CR, van Hensbergen Y, Luther S, Aghai Z, Rychnavská ZS, Slot M, Scherjon S, Kröger N, Ganser A, Weissinger EM, Goulmy E, Hambach L. Possible role of minor h antigens in the persistence of donor chimerism after stem cell transplantation; relevance for sustained leukemia remission. PLoS One 2015; 10:e0119595. [PMID: 25774796 PMCID: PMC4361395 DOI: 10.1371/journal.pone.0119595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 01/14/2015] [Indexed: 12/22/2022] Open
Abstract
Persistent complete donor chimerism is an important clinical indicator for remissions of hematological malignancies after HLA-matched allogeneic stem cell transplantation (SCT). However, the mechanisms mediating the persistence of complete donor chimerism are poorly understood. The frequent coincidence of complete donor chimerism with graft-versus-leukemia effects and graft-versus-host disease suggests that immune responses against minor histocompatibility antigens (mHags) are playing an important role in suppressing the host hematopoiesis after allogeneic SCT. Here, we investigated a possible relationship between donor immune responses against the hematopoiesis-restricted mHag HA-1 and the long-term kinetics of host hematopoietic chimerism in a cohort of 10 patients after allogeneic HLA-matched, HA-1 mismatched SCT. Functional HA-1 specific CTLs (HA-1 CTLs) were detectable in 6/10 patients lysing host-type hematopoietic cells in vitro. Presence of HA-1 CTLs in the peripheral blood coincided with low host hematopoiesis levels quantified by highly sensitive mHag specific PCR. Additionally, co-incubation of host type CD34+ cells with HA-1 CTLs isolated after allogeneic SCT prevented progenitor and cobblestone area forming cell growth in vitro and human hematopoietic engraftment in immunodeficient mice. Conversely, absence or loss of HA-1 CTLs mostly coincided with high host hematopoiesis levels and/or relapse. In summary, in this first study, presence of HA-1 CTLs paralleled low host hematopoiesis levels. This coincidence might be supported by the capacity of HA-1 CTLs isolated after allogeneic SCT to specifically eliminate host type hematopoietic stem/progenitor cells. Additional studies involving multiple mismatched mHags in more patients are required to confirm this novel characteristic of mHag CTLs as factor for the persistence of complete donor chimerism and leukemia remission after allogeneic SCT.
Collapse
Affiliation(s)
- Cornelis R. van der Torren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Yvette van Hensbergen
- Sanquin Blood Supply Foundation, Division of Research, Department of Transfusion Medicine, Leiden, The Netherlands
| | - Susanne Luther
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Zohara Aghai
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Zuzana Stachová Rychnavská
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Manon Slot
- Sanquin Blood Supply Foundation, Division of Research, Department of Transfusion Medicine, Leiden, The Netherlands
| | - Sicco Scherjon
- Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Nicolaus Kröger
- Department of Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Eva M. Weissinger
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Els Goulmy
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Lothar Hambach
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
11
|
Pessach I, Resnick I, Shimoni A, Nagler A. G-CSF-primed BM for allogeneic SCT: revisited. Bone Marrow Transplant 2015; 50:892-8. [DOI: 10.1038/bmt.2015.25] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/04/2015] [Accepted: 01/05/2015] [Indexed: 01/08/2023]
|
12
|
Relapse assessment following allogeneic SCT in patients with MDS and AML. Ann Hematol 2014; 93:1097-110. [PMID: 24671364 DOI: 10.1007/s00277-014-2046-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 02/25/2014] [Indexed: 10/25/2022]
Abstract
Options to pre-emptively treat impending relapse of myelodysplastic syndromes (MDS) and acute myeloid leukaemia (AML) after allogeneic haematopoietic stem cell transplantation (allo-SCT) continuously increase. In recent years, the spectrum of diagnostic methods and parameters to perform post-transplant monitoring in patients with AML and MDS has grown. Cytomorphology, histomorphology, and chimaerism analysis are the mainstay in any panel of post-transplant monitoring. This may be individually combined with multiparameter flow cytometry (MFC) for the detection of residual cells with a leukaemia phenotype and quantitative real-time polymerase chain reaction (RQ-PCR) to assess gene expression, e.g., of WT1 or the residual mutation load (e.g., in case of an NPM1 mutation). Data evaluating the aforementioned methods alone or in combination are discussed in this review with particular emphasis on data pointing towards their suitability to steer pre-emptive post-transplant interventions such as immunotherapy, chemotherapy or therapy with demethylating agents.
Collapse
|
13
|
Wayne AS, Giralt S, Kröger N, Bishop MR. Proceedings from the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: introduction. Biol Blood Marrow Transplant 2013; 19:1534-6. [PMID: 24035783 PMCID: PMC3880421 DOI: 10.1016/j.bbmt.2013.08.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 08/30/2013] [Indexed: 11/15/2022]
Abstract
Despite advances in hematopoietic stem cell transplantation (HSCT) for the treatment of hematologic malignancies, relapse remains the leading cause of death after transplant. Biologic and clinical investigations are needed to combat this primary cause of death after transplantation. The National Cancer Institute held international workshops in 2009 and 2012 to help address this problem. Three major initiatives for coordinated research were proposed: 1) To establish multicenter networks for basic, translational, epidemiologic and clinical research; 2) To establish a network of biorepositories for the collection of samples before and after HSCT to aid in laboratory and clinical studies; and 3) To refine, implement and study proposed definitions for disease-specific response and relapse and for monitoring of minimal residual disease. The workshop in 2012 also featured nine presentations, summaries of which follow in three manuscripts.
Collapse
Affiliation(s)
- Alan S Wayne
- Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, The Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California; Organizing Committee, National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation.
| | | | | | | |
Collapse
|
14
|
de Lima M, Porter DL, Battiwalla M, Bishop MR, Giralt SA, Hardy NM, Kröger N, Wayne AS, Schmid C. Proceedings from the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse After Hematopoietic Stem Cell Transplantation: part III. Prevention and treatment of relapse after allogeneic transplantation. Biol Blood Marrow Transplant 2013; 20:4-13. [PMID: 24018392 DOI: 10.1016/j.bbmt.2013.08.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 08/30/2013] [Indexed: 12/16/2022]
Abstract
In the Second Annual National Cancer Institute's Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation, the Scientific/Educational Session on the Prevention and Treatment of Relapse after Allogeneic Transplantation highlighted progress in developing new therapeutic approaches since the first relapse workshop. Recent insights that might provide a basis for the development of novel, practical clinical trials were emphasized, including utilization of newer agents, optimization of donor lymphocyte infusion (DLI), and investigation of novel cellular therapies. Dr. de Lima discussed pre-emptive and maintenance strategies to prevent relapse after transplantation, for example, recent promising results suggestive of enhanced graft-versus-tumor activity with hypomethylating agents. Dr. Schmid provided an overview of adjunctive strategies to improve cell therapy for relapse, including cytoreduction before DLI, combination of targeted agents with DLI, and considerations in use of second transplantations. Dr. Porter addressed strategies to enhance T cell function, including ex vivo activated T cells and T cell engineering, and immunomodulatory approaches to enhance T cell function in vivo, including exogenous cytokines and modulation of costimulatory pathways.
Collapse
Affiliation(s)
- Marcos de Lima
- Hematologic Malignancies and Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - David L Porter
- Blood and Marrow Transplantation, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Minoo Battiwalla
- Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Michael R Bishop
- Section of Hematology/Oncology, University of Chicago Medicine, Chicago, Illinois
| | - Sergio A Giralt
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Nancy M Hardy
- Experimental Transplantation Immunology Branch, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland.
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alan S Wayne
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland; Current: Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | |
Collapse
|
15
|
Shaffer BC, Modric M, Stetler-Stevenson M, Arthur DC, Steinberg SM, Liewehr DJ, Fowler DH, Gale RP, Bishop MR, Pavletic SZ. Rapid complete donor lymphoid chimerism and graft-versus-leukemia effect are important in early control of chronic lymphocytic leukemia. Exp Hematol 2013; 41:772-8. [PMID: 23689118 PMCID: PMC3769491 DOI: 10.1016/j.exphem.2013.04.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/12/2013] [Accepted: 04/18/2013] [Indexed: 01/29/2023]
Abstract
Eradication of minimal residual disease (MRD) after allotransplantation in persons with chronic lymphocytic leukemia (CLL) is associated with lower rates of relapse. Rapid engraftment of donor lymphocyte elements can contribute to MRD control, but it remains unclear whether this strategy will benefit patients. In this study, we report the incidence of MRD eradication and graft-versus-host disease (GvHD) in persons with rapid versus later donor T lymphocyte engraftment after lymphodepleting chemotherapy and reduced intensity conditioning (RIC) allotransplantation. Twenty-seven subjects received lymphodepleting chemotherapy to facilitate donor engraftment followed by fludarabine and cyclophosphamide RIC and a blood cell allograft. MRD was monitored by multicolor flow cytometry after transplantation. Complete donor T lymphoid chimerism (TLC) and myeloid chimerism (MC) were achieved in 25 subjects at a median of 28 days (range, 14-60 days) and 21 days (range, 14-180 days), respectively. Achieving complete donor TLC by day 14 versus day 28 or later correlated with occurrence of grade 2 or higher acute GvHD (90% [95% confidence interval {CI}, 78-100%] versus 35% [95% CI, 16-54%]; p = 0.014) and better control of MRD in the bone marrow at day 100, median 0% (range, 0-0.1%) versus 8.5% (range, 0-92%; p = 0.016). Among 11 persons with early donor TLC, none had progressive disease, and seven died of treatment -related mortality (TRM). In persons with later development of TLC, 8 of 16 had progressive disease and 2 died of TRM. Time to donor myeloid chimerism had no effect on outcomes. Rapid establishment of donor TLC resulted in more complete eradication of early MRD, but greater incidence of acute GvHD and TRM in persons with CLL undergoing RIC allotransplantation.
Collapse
Affiliation(s)
- Brian C Shaffer
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Minimal residual disease quantification using consensus primers and high-throughput IGH sequencing predicts post-transplant relapse in chronic lymphocytic leukemia. Leukemia 2013; 27:1659-65. [PMID: 23419792 PMCID: PMC3740398 DOI: 10.1038/leu.2013.52] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 02/06/2013] [Accepted: 02/11/2013] [Indexed: 12/21/2022]
Abstract
Quantification of minimal residual disease (MRD) following allogeneic hematopoietic cell transplantation (allo-HCT) predicts post-transplant relapse in patients with chronic lymphocytic leukemia (CLL). We utilized an MRD-quantification method that amplifies immunoglobulin heavy chain (IGH) loci using consensus V and J segment primers followed by high-throughput sequencing (HTS), enabling quantification with a detection limit of one CLL cell per million mononuclear cells. Using this IGH–HTS approach, we analyzed MRD patterns in over 400 samples from 40 CLL patients who underwent reduced-intensity allo-HCT. Nine patients relapsed within 12 months post-HCT. Of the 31 patients in remission at 12 months post-HCT, disease-free survival was 86% in patients with MRD <10−4 and 20% in those with MRD ⩾10−4 (relapse hazard ratio (HR) 9.0; 95% confidence interval (CI) 2.5–32; P<0.0001), with median follow-up of 36 months. Additionally, MRD predicted relapse at other time points, including 9, 18 and 24 months post-HCT. MRD doubling time <12 months with disease burden ⩾10−5 was associated with relapse within 12 months of MRD assessment in 50% of patients, and within 24 months in 90% of patients. This IGH–HTS method may facilitate routine MRD quantification in clinical trials.
Collapse
|
17
|
Abstract
In the study reported in this issue of Blood by Leung et al, detectable minimal residual disease (MRD) before hematopoietic stem cell transplantation (HSCT) was found to be prognostic for outcome, but did not prevent cure for children with acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML).
Collapse
|
18
|
Nagler A, Labopin M, Shimoni A, Mufti GJ, Cornelissen JJ, Blaise D, Janssen JJWM, Milpied N, Vindelov L, Petersen E, Gribben J, Bacigalupo A, Malm C, Niederwieser D, Socié GJ, Arnold R, Brown P, Goker H, Rocha V, Mohty M. Mobilized peripheral blood stem cells compared with bone marrow from HLA-identical siblings for reduced-intensity conditioning transplantation in acute myeloid leukemia in complete remission: a retrospective analysis from the Acute Leukemia Working Party of EBMT. Eur J Haematol 2012; 89:206-13. [PMID: 22650267 DOI: 10.1111/j.1600-0609.2012.01811.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2012] [Indexed: 12/20/2022]
Abstract
Reduced-intensity conditioning (RIC)-alloSCT is increasingly used for acute myelogenous leukemia. Limited data are available for the comparison of peripheral blood stem cells with bone marrow for RIC-alloSCT. We used the European Group for Blood and Marrow Transplantation (EBMT) ALWP data to compare the outcome of mobilized peripheral blood stem cells (PBSC) (n = 1430) vs. bone marrow (BM) (n = 107) for acute myelogenous leukemia (AML) patients with complete remission that underwent RIC-alloSCT from compatible sibling donors. The leukemia features, the disease status, and the time from diagnosis were similar between the two groups. Engraftment was achieved in 99% and 93% in the PBSC and BM groups, respectively (P < 0.0001). The day of engraftment was significantly earlier for the PBSC vs. the BM group, 15 (1-59) and 19 (5-69), respectively (P < 0.001). Acute GVHD, severe GVHD (grade III-IV) and chronic GVHD did not differ between the groups. leukemia-free survival (LFS), relapse, and non-relapsed mortality (NRM) were 51 ± 2%, 32 ± 1%, and 17 ± 1% vs. 50 ± 6%, 38 ± 6%, and 12 ± 3% for the PBSC and BM groups, respectively. Our results indicate faster engraftment, but no difference in GVHD, LFS, relapse, and NRM when comparing PBSC to BM grafts from sibling donors following RIC conditioning. This is the first study comparing PBSC to BM grafts in the RIC setting, analyzing a homogeneous population of patients with AML in remission. Whether PBSC should be preferred for advanced phases of the disease, where the outcome is dominated by relapse incidences, needs further investigation.
Collapse
Affiliation(s)
- Arnon Nagler
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Israel.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Abstract
Allogeneic hematopoietic stem cell transplantation (SCT) is well accepted as a curative treatment approach for younger patients with myelodysplastic syndrome (MDS) and has become one of the most frequent indications for allogeneic SCT as reported to the Center for International Blood and Marrow Transplant Research. However, MDS patients are usually elderly with a median age of approximately 75 years at diagnosis. Large register studies have confirmed the feasibility of the procedure in elderly MDS patients; and in the register of the European Group for Blood and Marrow Transplantation, one-third of the allogeneic transplant procedures for MDS were performed in 2010 in patients older than 60 years. Despite its curative potential, its role in the treatment of elderly MDS patients is less defined. Because of the inherent complications of the transplantation leading to treatment-related mortality and the risk of relapse, a careful calculation of the benefit for each patient is mandatory, taking into account comorbidities, disease status, donor selection, and effective nontransplant therapies. Prospective multicenter studies are needed to define optimal intensity of the conditioning regimen, timing of transplantation within a treatment algorithm, including drug-based therapies, and posttransplant strategies to reduce the risk of relapse.
Collapse
|
20
|
Toze CL, Dalal CB, Nevill TJ, Gillan TL, Abou Mourad YR, Barnett MJ, Broady RC, Forrest DL, Hogge DE, Nantel SH, Power MM, Song KW, Sutherland HJ, Smith CA, Narayanan S, Young SS, Connors JM, Shepherd JD. Allogeneic haematopoietic stem cell transplantation for chronic lymphocytic leukaemia: outcome in a 20-year cohort. Br J Haematol 2012; 158:174-185. [DOI: 10.1111/j.1365-2141.2012.09170.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 03/06/2012] [Indexed: 11/30/2022]
Affiliation(s)
- Cynthia L. Toze
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Chinmay B. Dalal
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Thomas J. Nevill
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Tanya L. Gillan
- Cytogenetics Laboratory; Department of Pathology and Laboratory Medicine; Vancouver General Hospital and University of British Columbia; Vancouver BC
| | - Yasser R. Abou Mourad
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Michael J. Barnett
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Raewyn C. Broady
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Donna L. Forrest
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Donna E. Hogge
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Stephen H. Nantel
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Maryse M. Power
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Kevin W. Song
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Heather J. Sutherland
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Clayton A. Smith
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Sujaatha Narayanan
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| | - Sean S. Young
- Cancer Genetics Laboratory; Department of Pathology and Laboratory Medicine; BC Cancer Agency and University of British Columbia; Vancouver BC
| | - Joseph M. Connors
- Division of Medical Oncology; BC Cancer Agency; Centre for Lymphoid Cancer and University of British Columbia; Vancouver BC Canada
| | - John D. Shepherd
- Division of Hematology; Leukemia/BMT Program of British Columbia; Vancouver General Hospital; BC Cancer Agency; University of British Columbia; Vancouver BC
| |
Collapse
|
21
|
Current world literature. Curr Opin Oncol 2011; 23:700-9. [PMID: 21993416 DOI: 10.1097/cco.0b013e32834d384a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Abstract
PURPOSE OF REVIEW Relapse has become the leading cause of death following allogeneic hematopoietic stem cell transplantation (HSCT). Despite improved understanding of the biology that underlies the graft-versus-leukemia/tumor effect the relapse rate did not decrease over the past 20 years. In general, prognosis is poor for patients who relapsed to an allograft since effective treatment options are limited. Here, we review the available and upcoming treatment approaches for relapse. RECENT FINDINGS Treatment of relapse after allogeneic HSCT has been rarely investigated systematically and results differ substantially from diseases. Withdrawal of immunosuppressive medication, donor lymphocyte infusions with or without chemotherapy and/or second allogeneic HSCT are the most used options. New specific cellular approaches such as disease-specific T-cells, alloreactive natural killer cells or vaccination strategies are under investigation. Novel agents such as tyrosine-kinase inhibitors, hypomethylating agents, monoclonal antibodies, immunomodulating drugs, or proteasome-inhibitors either alone or in combination with adoptive immunotherapy are upcoming promising options, but valid data are lacking so far. SUMMARY With some exceptions (chronic myeloid leukemia), treatment options for patients who relapse are limited. The results are poor and the majority of patients ultimately die of their disease. More effort and research is needed to prevent and treat relapse after allogeneic HSCT.
Collapse
|
23
|
Roddie C, Peggs KS. Donor lymphocyte infusion following allogeneic hematopoietic stem cell transplantation. Expert Opin Biol Ther 2011; 11:473-87. [PMID: 21269237 DOI: 10.1517/14712598.2011.554811] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Allogeneic hematopoietic stem cell transplantation (SCT) is the treatment of choice for many malignant hematological disorders. Following recent improvements in non-relapse-related mortality rates, relapse has become the commonest cause of treatment failure. Infusion of donor lymphocytes can potentially enhance immune-mediated antitumor activity and offers a salvage option for some patients. This paper reviews the current literature on the efficacy of this therapeutic strategy. AREAS COVERED The biology of adoptive cellular therapy with allogeneic immune cells to treat relapse across a spectrum of diseases in both the full intensity and reduced intensity hematopoietic SCT settings is explored. The review discusses the current limitations of the approach and reviews several new experimental strategies which aim to segregate the desired graft-versus-tumor effect from the deleterious effects of more widespread graft-versus-host reactivity. EXPERT OPINION Durable responses to DLI have been noted in chronic myeloid leukemia and responses have also been described in acute leukemia, multiple myeloma and chronic lymphoproliferative disorders. The new challenge in transplantation is to optimize DLI therapy in order to further improve patient outcomes.
Collapse
Affiliation(s)
- Claire Roddie
- UCL Cancer Institute, Department of Haematology, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | | |
Collapse
|
24
|
Bishop MR, Alyea EP, Cairo MS, Falkenburg JHF, June CH, Kröger N, Little RF, Miller JS, Pavletic SZ, Porter DL, Riddell SR, van Besien K, Wayne AS, Weisdorf DJ, Wu RS, Giralt S. National Cancer Institute's First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: summary and recommendations from the organizing committee. Biol Blood Marrow Transplant 2011; 17:443-54. [PMID: 21224011 DOI: 10.1016/j.bbmt.2010.12.713] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 12/26/2010] [Indexed: 01/01/2023]
Abstract
The National Cancer Institute's First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation was organized and convened to identify, prioritize, and coordinate future research activities related to relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Each of the Workshop's 6 Working Committees has published individual reports of ongoing basic, translational, and clinical research and recommended areas for future research related to the areas of relapse biology, epidemiology, prevention, and treatment. This document summarizes each committee's recommendations and suggests 3 major initiatives for a coordinated research effort to address the problem of relapse after allo-HSCT: (1) to establish multicenter correlative and clinical trial networks for basic/translational, epidemiologic, and clinical research; (2) to establish a network of biorepositories for the collection of samples before and after allo-HSCT to aid in laboratory and clinical studies; and (3) to further refine, implement, and study the Workshop-proposed definitions for disease-specific response and relapse and recommendations for monitoring of minimal residual disease. These recommendations, in coordination with ongoing research initiatives and transplantation organizations, provide a research framework to rapidly and efficiently address the significant problem of relapse after allo-HSCT.
Collapse
Affiliation(s)
- Michael R Bishop
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Porter DL. Allogeneic immunotherapy to optimize the graft-versus-tumor effect: concepts and controversies. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2011; 2011:292-298. [PMID: 22160048 DOI: 10.1182/asheducation-2011.1.292] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Allogeneic stem cell transplantation (SCT) can be considered the most successful method of adoptive immunotherapy of cancer. It is successful in part because of the potent graft-versus-tumor (GVT) effects of the donor graft, which are independent of the conditioning regimen. This potent GVT reaction can be harnessed in some cases to treat patients who relapse after allogeneic SCT with the use of donor leukocyte infusions (DLIs). This has led to the rapid development of reduced-intensity conditioning (RIC) regimens for allogeneic SCT, an approach that relies primarily on GVT activity. However, the effects of GVT have clear disease specificity and remain associated with significant GVHD. Optimization of GVT induction will require a better understanding of the important target antigens and effector cells, as well as the development of methods that enhance GVT reactivity without excessive GVHD. The appropriate clinical setting and timing for GVT induction need to be defined more clearly, but ultimately, the immunologic control of cancer through allogeneic adoptive immunotherapy represents one of the most potent and promising therapeutic strategies for patients with hematologic malignancies.
Collapse
Affiliation(s)
- David L Porter
- Division of Hematology-Oncology, Blood and Marrow Transplant Program, University of Pennsylvania Medical Center, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Porter DL, Alyea EP, Antin JH, DeLima M, Estey E, Falkenburg JHF, Hardy N, Kroeger N, Leis J, Levine J, Maloney DG, Peggs K, Rowe JM, Wayne AS, Giralt S, Bishop MR, van Besien K. NCI First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: Report from the Committee on Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2010; 16:1467-503. [PMID: 20699125 PMCID: PMC2955517 DOI: 10.1016/j.bbmt.2010.08.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 08/03/2010] [Indexed: 12/31/2022]
Abstract
Relapse is a major cause of treatment failure after allogeneic hematopoietic stem cell transplantation (alloHSCT). Treatment options for relapse have been inadequate, and the majority of patients ultimately die of their disease. There is no standard approach to treating relapse after alloHSCT. Withdrawal of immune suppression and donor lymphocyte infusions are commonly used for all diseases; although these interventions are remarkably effective for relapsed chronic myelogenous leukemia, they have limited efficacy in other hematologic malignancies. Conventional and novel chemotherapy, monoclonal antibody therapy, targeted therapies, and second transplants have been utilized in a variety of relapsed diseases, but reports on these therapies are generally anecdotal and retrospective. As such, there is an immediate need for well-designed, disease-specific trials for treatment of relapse after alloHSCT. This report summarizes current treatment options under investigation for relapse after alloHSCT in a disease-specific manner. In addition, recommendations are provided for specific areas of research necessary in the treatment of relapse after alloHSCT.
Collapse
MESH Headings
- Hematologic Neoplasms/therapy
- Hematopoietic Stem Cell Transplantation
- Hodgkin Disease/therapy
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myeloid, Acute/therapy
- Lymphocyte Transfusion
- Lymphoma, Non-Hodgkin
- Multiple Myeloma/therapy
- Neoplasm Recurrence, Local/therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Recurrence
- Transplantation, Homologous
- Treatment Failure
Collapse
Affiliation(s)
- David L Porter
- University of Pennsylvania Medical Center, Philadelphia, 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Minimal residual disease following allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2010; 17:S94-100. [PMID: 21047560 DOI: 10.1016/j.bbmt.2010.10.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
|