201
|
Dillon LW, Hayati S, Roloff GW, Tunc I, Pirooznia M, Mitrofanova A, Hourigan CS. Targeted RNA-sequencing for the quantification of measurable residual disease in acute myeloid leukemia. Haematologica 2018; 104:297-304. [PMID: 30171026 PMCID: PMC6355494 DOI: 10.3324/haematol.2018.203133] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022] Open
Abstract
Great effort is spent on developing therapies to improve the dire outcomes of those diagnosed with acute myeloid leukemia. The methods for quantifying response to therapeutic intervention have however lacked sensitivity. Patients achieving a complete remission as defined by conventional cytomorphological methods therefore remain at risk of subsequent relapse due to disease persistence. Improved risk stratification is possible based on tests designed to detect this residual leukemic burden (measurable residual disease). However, acute myeloid leukemia is a genetically diverse set of diseases, which has made it difficult to develop a single, highly reproducible, and sensitive assay for measurable residual disease. Here we present the development of a digital targeted RNA-sequencing-based approach designed to overcome these limitations by detecting all newly approved European LeukemiaNet molecular targets for measurable residual disease in acute myeloid leukemia in a single standardized assay. Iterative modifications and novel bioinformatics approaches resulted in a greater than 100-fold increase in performance compared with commercially available targeted RNA-sequencing approaches and a limit of detection as low as one leukemic cell in 100,000 cells measured, which is comparable to quantitative polymerase chain reaction analysis, the current gold standard for the detection of measurable residual disease. This assay, which can be customized and expanded, is the first demonstrated use of high-sensitivity RNA-sequencing for measurable residual disease detection in acute myeloid leukemia and could serve as a broadly applicable standardized tool.
Collapse
Affiliation(s)
- Laura W Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Sheida Hayati
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.,Department of Health Informatics, Rutgers School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ
| | - Gregory W Roloff
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ilker Tunc
- Bioinformatics and Computational Biology Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Antonina Mitrofanova
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
202
|
Canaani J, Labopin M, Huang XJ, Ciceri F, Van Lint MT, Bruno B, Santarone S, Diez-Martin JL, Blaise D, Chiusolo P, Wu D, Mohty M, Nagler A. Minimal residual disease status predicts outcome of acute myeloid leukaemia patients undergoing T-cell replete haploidentical transplantation. An analysis from the Acute Leukaemia Working Party (ALWP) of the European Society for Blood and Marrow Transplan. Br J Haematol 2018; 183:411-420. [DOI: 10.1111/bjh.15540] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/29/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Jonathan Canaani
- Haematology Division; Chaim Sheba Medical Centre; Tel Aviv University; Tel Hashomer Israel
| | - Myriam Labopin
- EBMT Paris study office/CEREST-TC; Hopital Saint-Antoine; Paris France
| | - Xiao J. Huang
- Haematology; Peking University People′s Hospital; Peking China
| | - Fabio Ciceri
- Haematology and Bone Marrow Transplantation Unit; San Raffaele Scientific Institute; Milan Italy
| | | | | | | | - José L. Diez-Martin
- Hospital GU Gregorio Marañon; Instituto de investigación sanitaria Gregorio Marañon; Facultad de Medicina; Universidad Complutense; Madrid Spain
| | - Didier Blaise
- Haematology Department; Institut Paoli Calmettes; Marseille France
| | - Patrizia Chiusolo
- Department of Haematology; Universita Cattolica Sacro Cuore; Rome Italy
| | - Depei Wu
- Department of Haematology; the First Affiliated Hospital of Soochow University; Soochow China
| | - Mohamad Mohty
- EBMT Paris study office/CEREST-TC; Hopital Saint-Antoine; Paris France
| | - Arnon Nagler
- Haematology Division; Chaim Sheba Medical Centre; Tel Aviv University; Tel Hashomer Israel
- EBMT Paris study office/CEREST-TC; Hopital Saint-Antoine; Paris France
| |
Collapse
|
203
|
Liu J, Liu YR, Wang YZ, Han W, Chen H, Chen Y, Wang JZ, Mo XD, Zhang YY, Yan CH, Sun YQ, Chen YY, Wang Y, Xu LP, Zhang XH, Liu KY, Huang XJ, Chang YJ. [The comparison of predicting clinical outcomes between immunolophenotype and hematological complete remission before human leukocyte antigen-matched sibling donor transplantation in acute myeloid leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:617-623. [PMID: 30180459 PMCID: PMC7342848 DOI: 10.3760/cma.j.issn.0253-2727.2018.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Indexed: 11/18/2022]
Abstract
Objective: To assess the prognostic significance of immunophenotype complete remission (ICR) and hematological complete remission (HCR) before human-leukocyte antigen (HLA)-matched sibling donor transplantation (MSDT) in acute myeloid leukemia (AML) patients. Methods: A cohort of 182 AML (non-APL) patients undergoing MSDT in HCR was retrospectively studied [including complete remission with ANC and PLT recovery (CR), CR with incomplete PLT recovery (CRp), CR with inconplete ANC and PLT recovery (CRi)]; ICR was determined as undetective minimal resudial disease (MRD) by multi-parameter flow cytometer. Results: ①Of the 182 patients, 97 were male, 85 female, and the median age was 41(4-62) years. ②The CR and CRi+CRp rates were 80.8% (147/182) and 19.2%(35/182), respectively; The 4-year cumulative incidence of relapse[CIR, (11.0±4.3)% vs (16.0±7.1)%, χ(2)=0.274, P=0.600], non-relapse mortality[NRM, (14.0±4.3)% vs (9.0±6.3)%, χ(2)=0.913, P=0.339], leukemia-free survival[LFS, (75.0±5.1)% vs (75.0±8.3)%, χ(2)=0.256, P=0.613], and overall survial [OS, (77.0±5.2)% vs (80.0±8.1)%, χ(2)=0.140, P=0.708] were comparable between the CRp+CRi and CR groups. ③Compared with the non-ICR group (n=35), the ICR group (n=147) showed lower 4-year CIR [(11.3±3.4) % vs (55.2±8.8) %, χ(2)=32.687, P<0.001], better 4-year LFS [(76.2±4.7)% vs (32.8±8.7)%, χ(2)=26.234, P<0.001] and OS[(79.0±4.7)% vs (39.0±9.1)%, χ(2)=25.253, P<0.001], and comparable NRM[(12.5±4.1)% vs (12.0±7.1)%, χ(2)=1.002, P=0.656]. ④Mulitvariate analysis confirmed the independent prognostic value of ICR in lower CIR [HR=11.026(95%CI 4.685-25.949), P<0.001], higher LFS [HR=5.785 (95% CI 2.974-11.254), P<0.001] and OS[HR=5.578 (95% CI 2.575-27.565), P<0.001]. Conclusion: The results indicated that ICR instead of HCR pre-transplantation had a significant prognostic value in AML patients undergoing MSDT.
Collapse
Affiliation(s)
- J Liu
- Institute of Hematology, Peoples' Hospital, Peking University, Beijing 100044, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Warlick ED, DeFor TE, Bejanyan N, Holtan S, MacMillan M, Blazar BR, Dusenbery K, Arora M, Bachanova V, Cooley S, Lazaryan A, McGlave P, Miller JS, Rashidi A, Slungaard A, Vercellotti G, Ustun C, Brunsein C, Weisdorf D. Reduced-Intensity Conditioning Followed by Related and Unrelated Allografts for Hematologic Malignancies: Expanded Analysis and Long-Term Follow-Up. Biol Blood Marrow Transplant 2018; 25:56-62. [PMID: 30077015 DOI: 10.1016/j.bbmt.2018.07.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/23/2018] [Indexed: 01/27/2023]
Abstract
Reduced-intensity conditioning (RIC) extends the curative potential of allogeneic hematopoietic cell transplantation (HCT) to patients with hematologic malignancies unable to withstand myeloablative conditioning. We prospectively analyzed the outcomes of 292 consecutive patients, median age 58 years (range, 19 to 75) with hematologic malignancies treated with a uniform RIC regimen of cyclophosphamide, fludarabine, and total body irradiation (200 cGy) with or without antithymocyte globulin and cyclosporine and mycophenolate mofetil graft-versus-host disease (GVHD) prophylaxis followed by allogeneic HCT at the University of Minnesota from 2002 to 6. Probability of 5-year overall survival was 78% for patients with indolent non-Hodgkin lymphoma, 53% for chronic myelogenous leukemia, 55% for Hodgkin lymphoma, 40% for acute myelogenous leukemia, 37% for myelodysplastic syndrome, 29% for myeloma, and 14% for myeloproliferative neoplasms. Corresponding outcomes for relapse were 0%, 13%, 53%, 37%, 39%, 75%, and 29%, respectively. Disease risk index (DRI) predicted both survival and relapse with superior survival (64%) and lowest relapse (16%) in those with low risk score compared with 24% survival and 57% relapse in those with high/very-high risk scores. Recipient cytomegalovirus (CMV)-positive serostatus was protective from relapse with the lowest rates in those also receiving a CMV-positive donor graft (29%). The cumulative incidence of 2-year nonrelapse mortality was 26% and was lowest in those receiving a matched sibling graft at 21%, with low (21%) or intermediate (18%) HCT-specific comorbidity index, and was similar across age groups. The incidence of grades II to IV acute GVHD was 43% and grades III to IV 27%; the highest rates were found in those receiving an unrelated donor (URD) peripheral blood stem cell (PBSC) graft, at 50%. Chronic GVHD at 1 year was 36%. Future approaches incorporating alternative GVHD prophylaxis, particularly for URD PBSC grafts, and targeted post-transplant antineoplastic therapies for those with high DRI are indicated to improve these outcomes.
Collapse
Affiliation(s)
- Erica Dahl Warlick
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota.
| | - Todd E DeFor
- Biostatistics and Informatics Core, Masonic Cancer Center, Minneapolis, Minnesota
| | - Nelli Bejanyan
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Shernan Holtan
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Margaret MacMillan
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Bruce R Blazar
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Kathryn Dusenbery
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Mukta Arora
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Veronika Bachanova
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Sarah Cooley
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Aleksandr Lazaryan
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Philip McGlave
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey S Miller
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Armin Rashidi
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Arne Slungaard
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Gregory Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Celalettin Ustun
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Claudio Brunsein
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Daniel Weisdorf
- Department of Medicine, Division of Hematology, Oncology and Transplant, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
205
|
Martínez-Laperche C, Buces E, Aguilera-Morillo MC, Picornell A, González-Rivera M, Lillo R, Santos N, Martín-Antonio B, Guillem V, Nieto JB, González M, de la Cámara R, Brunet S, Jiménez-Velasco A, Espigado I, Vallejo C, Sampol A, Bellón JM, Serrano D, Kwon M, Gayoso J, Balsalobre P, Urbano-Izpizua Á, Solano C, Gallardo D, Díez-Martín JL, Romo J, Buño I. A novel predictive approach for GVHD after allogeneic SCT based on clinical variables and cytokine gene polymorphisms. Blood Adv 2018; 2:1719-1737. [PMID: 30030270 PMCID: PMC6058238 DOI: 10.1182/bloodadvances.2017011502] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
Despite considerable advances in our understanding of the pathophysiology of graft-versus-host disease (GVHD), its prediction remains unresolved and depends mainly on clinical data. The aim of this study is to build a predictive model based on clinical variables and cytokine gene polymorphism for predicting acute GVHD (aGVHD) and chronic GVHD (cGVHD) from the analysis of a large cohort of HLA-identical sibling donor allogeneic stem cell transplant (allo-SCT) patients. A total of 25 SNPs in 12 cytokine genes were evaluated in 509 patients. Data were analyzed using a linear regression model and the least absolute shrinkage and selection operator (LASSO). The statistical model was constructed by randomly selecting 85% of cases (training set), and the predictive ability was confirmed based on the remaining 15% of cases (test set). Models including clinical and genetic variables (CG-M) predicted severe aGVHD significantly better than models including only clinical variables (C-M) or only genetic variables (G-M). For grades 3-4 aGVHD, the correct classification rates (CCR1) were: 100% for CG-M, 88% for G-M, and 50% for C-M. On the other hand, CG-M and G-M predicted extensive cGVHD better than C-M (CCR1: 80% vs. 66.7%, respectively). A risk score was calculated based on LASSO multivariate analyses. It was able to correctly stratify patients who developed grades 3-4 aGVHD (P < .001) and extensive cGVHD (P < .001). The novel predictive models proposed here improve the prediction of severe GVHD after allo-SCT. This approach could facilitate personalized risk-adapted clinical management of patients undergoing allo-SCT.
Collapse
Affiliation(s)
- Carolina Martínez-Laperche
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Elena Buces
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | - Antoni Picornell
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Oncology and
| | - Milagros González-Rivera
- Department of Oncology and
- DNA Sequencing and Genotyping Core Facility, H.G.U. Gregorio Marañón, Madrid, Spain
| | - Rosa Lillo
- Department of Statistics, Universidad Carlos III de Madrid, Madrid, Spain
| | - Nazly Santos
- Department of Hematology, Instituto Catalán de Oncología Hospital Josep Trueta, Girona, Spain
| | - Beatriz Martín-Antonio
- Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Vicent Guillem
- Department of Hematology, Hospital Clínico de Valencia, Valencia, Spain
| | - José B Nieto
- Department of Hematology, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Marcos González
- Department of Hematology, Hospital de Salamanca, Salamanca, Spain
| | - Rafael de la Cámara
- Department of Hematology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Salut Brunet
- Department of Haematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Ildefonso Espigado
- Department of Hematology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Carlos Vallejo
- Department of Hematology, Hospital Central de Asturias, Oviedo, Spain
| | - Antonia Sampol
- Department of Hematology, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - José María Bellón
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - David Serrano
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Mi Kwon
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Jorge Gayoso
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Pascual Balsalobre
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Álvaro Urbano-Izpizua
- Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Carlos Solano
- Department of Hematology, Hospital Clínico de Valencia, Valencia, Spain
| | - David Gallardo
- Department of Hematology, Instituto Catalán de Oncología Hospital Josep Trueta, Girona, Spain
| | - José Luis Díez-Martín
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Medicine, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and
| | - Juan Romo
- Department of Statistics, Universidad Carlos III de Madrid, Madrid, Spain
| | | |
Collapse
|
206
|
Molecular Minimal Residual Disease Testing in Acute Myeloid Leukemia: A Review for the Practicing Clinician. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:636-647. [PMID: 30006258 DOI: 10.1016/j.clml.2018.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 11/21/2022]
Abstract
Minimal residual disease (MRD) testing in acute myeloid leukemia is increasingly being used to assess treatment response and stratify the risk of relapse for individual patients. Molecular methods for MRD testing began with PCR-based assays for individual recurrent mutations. To date, there is robust evidence for testing NPM1, CBFB-MYH11, and RUNX1/RUNXT1 mutations using this approach, though the best timing and threshold level for each mutation varies. More recent approaches have been with PCR-based multigene panels, occasionally combined with flow cytometric techniques, and next-generation sequencing techniques. This review outlines the various techniques used in molecular approaches to MRD, the evidence behind individual mutation testing, and the novel approaches for evaluating multigene MRD so that clinicians can understand and incorporate these evaluations into their practice.
Collapse
|
207
|
Buccisano F, Hourigan CS, Walter RB. The Prognostic Significance of Measurable ("Minimal") Residual Disease in Acute Myeloid Leukemia. Curr Hematol Malig Rep 2018; 12:547-556. [PMID: 29027628 DOI: 10.1007/s11899-017-0420-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review was to evaluate recent literature on detection methodologies for, and prognostic significance of, measurable ("minimal") residual disease (MRD) in acute myeloid leukemia (AML). RECENT FINDINGS There is no "one-fits-all" approach to MRD testing in AML. Most exploited to date are methods relying on immunophenotypic aberrancies (identified via multiparameter flow cytometry) or genetic abnormalities (identified via PCR-based assays). Current methods have important shortcomings, including the lack of assay platform standardization/harmonization across laboratories. In parallel to refinements of existing technologies and data analysis/interpretation, new methodologies (e.g., next-generation sequencing-based assays) are emerging that eventually may complement or replace existing ones. This dynamic evolution of MRD testing has complicated comparisons between individual studies. Nonetheless, an ever-growing body of data demonstrates that a positive MRD test at various time points throughout chemotherapy and hematopoietic cell transplantation identifies patients at particularly high risks of disease recurrence and short survival even after adjustment for other risk factors.
Collapse
Affiliation(s)
- Francesco Buccisano
- Department of Biomedicine and Prevention, Hematology, University Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| | - Christopher S Hourigan
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.,Department of Epidemiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
208
|
Selim AG, Moore AS. Molecular Minimal Residual Disease Monitoring in Acute Myeloid Leukemia. J Mol Diagn 2018; 20:389-397. [DOI: 10.1016/j.jmoldx.2018.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/22/2018] [Accepted: 03/27/2018] [Indexed: 01/22/2023] Open
|
209
|
Buccisano F, Dillon R, Freeman SD, Venditti A. Role of Minimal (Measurable) Residual Disease Assessment in Older Patients with Acute Myeloid Leukemia. Cancers (Basel) 2018; 10:cancers10070215. [PMID: 29949858 PMCID: PMC6070940 DOI: 10.3390/cancers10070215] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/18/2018] [Accepted: 06/18/2018] [Indexed: 12/22/2022] Open
Abstract
Minimal (or measurable) residual (MRD) disease provides a biomarker of response quality for which there is robust validation in the context of modern intensive treatment for younger patients with Acute Myeloid Leukemia (AML). Nevertheless, it remains a relatively unexplored area in older patients with AML. The lack of progress in this field can be attributed to two main reasons. First, physicians have a general reluctance to submitting older adults to intensive chemotherapy due to their frailty and to the unfavourable biological disease profile predicting a poor outcome following conventional chemotherapy. Second, with the increasing use of low-intensity therapies (i.e., hypomethylating agents) differing from conventional drugs in mechanism of action and dynamics of response, there has been concomitant skepticism that these schedules can produce deep hematological responses. Furthermore, age dependent differences in disease biology also contribute to uncertainty on the prognostic/predictive impact in older adults of certain genetic abnormalities including those validated for MRD monitoring in younger patients. This review examines the evidence for the role of MRD as a prognosticator in older AML, together with the possible pitfalls of MRD evaluation in older age.
Collapse
Affiliation(s)
- Francesco Buccisano
- Hematology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London SE1 9RT, UK.
| | - Sylvie D Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK.
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
| |
Collapse
|
210
|
DeStefano CB, Hourigan CS. Personalizing initial therapy in acute myeloid leukemia: incorporating novel agents into clinical practice. Ther Adv Hematol 2018; 9:109-121. [PMID: 29713444 DOI: 10.1177/2040620718761778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
While the past decade has seen a revolution in understanding of the genetic and molecular etiology of the disease, in clinical practice, initial therapy for acute myeloid leukemia (AML) patients has been a relatively straightforward choice between intensive combination cytotoxic induction therapy as used for decades or less-intensive hypomethylating therapy. The year 2017, however, witnessed US Food and Drug Administration approvals of midostaurin, enasidenib, gemtuzumab ozogamicin and CPX-351 for AML patients, with many other promising agents currently in clinical trials. This review discusses these options, highlights unanswered questions regarding optimal combinations and proposes some suggested approaches for the personalization of initial therapy for AML patients.
Collapse
Affiliation(s)
- Christin B DeStefano
- Laboratory of Myeloid Malignancies, National Institutes of Health, Bethesda, MD, USA Department of Hematology, MedStar Washington Cancer Institute, Washington, DC, USA
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5130, 10 Center Drive, Bethesda, MD 20814-1476, USA
| |
Collapse
|
211
|
Schuurhuis GJ, Heuser M, Freeman S, Béné MC, Buccisano F, Cloos J, Grimwade D, Haferlach T, Hills RK, Hourigan CS, Jorgensen JL, Kern W, Lacombe F, Maurillo L, Preudhomme C, van der Reijden BA, Thiede C, Venditti A, Vyas P, Wood BL, Walter RB, Döhner K, Roboz GJ, Ossenkoppele GJ. Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party. Blood 2018; 131:1275-1291. [PMID: 29330221 PMCID: PMC5865231 DOI: 10.1182/blood-2017-09-801498] [Citation(s) in RCA: 792] [Impact Index Per Article: 113.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022] Open
Abstract
Measurable residual disease (MRD; previously termed minimal residual disease) is an independent, postdiagnosis, prognostic indicator in acute myeloid leukemia (AML) that is important for risk stratification and treatment planning, in conjunction with other well-established clinical, cytogenetic, and molecular data assessed at diagnosis. MRD can be evaluated using a variety of multiparameter flow cytometry and molecular protocols, but, to date, these approaches have not been qualitatively or quantitatively standardized, making their use in clinical practice challenging. The objective of this work was to identify key clinical and scientific issues in the measurement and application of MRD in AML, to achieve consensus on these issues, and to provide guidelines for the current and future use of MRD in clinical practice. The work was accomplished over 2 years, during 4 meetings by a specially designated MRD Working Party of the European LeukemiaNet. The group included 24 faculty with expertise in AML hematopathology, molecular diagnostics, clinical trials, and clinical medicine, from 19 institutions in Europe and the United States.
Collapse
Affiliation(s)
- Gerrit J Schuurhuis
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Sylvie Freeman
- Department of Clinical Immunology, Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Francesco Buccisano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pediatric Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - David Grimwade
- Division of Genetics & Molecular Medicine, King's College, London, United Kingdom
| | | | - Robert K Hills
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | | | - Jeffrey L Jorgensen
- Division of Pathology/Laboratory Medicine, Department of Hematopathology, MD Anderson Cancer Center, Houston, TX
| | | | - Francis Lacombe
- Flow Cytometry Platform, University Hospital, Bordeaux, France
| | - Luca Maurillo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Claude Preudhomme
- Center of Pathology, Laboratory of Hematology, University Hospital of Lille, Lille, France
| | - Bert A van der Reijden
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian Thiede
- Universitätsklinikum Carl Gustav Garus an der Technischen Universität Dresden, Dresden, Germany
| | - Adriano Venditti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Paresh Vyas
- Medical Research Council Molecular Haematology Unit, Oxford Centre for Haematology, University of Oxford and Oxford University Hospitals National Health Service Trust, Oxford, United Kingdom
| | - Brent L Wood
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Laboratory Medicine and
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany; and
| | - Gail J Roboz
- Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY
| | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
212
|
Buccisano F, Maurillo L, Del Principe MI, Di Veroli A, De Bellis E, Biagi A, Zizzari A, Rossi V, Rapisarda V, Amadori S, Voso MT, Lo-Coco F, Arcese W, Venditti A. Minimal residual disease as a biomarker for outcome prediction and therapy optimization in acute myeloid leukemia. Expert Rev Hematol 2018; 11:307-313. [PMID: 29495904 DOI: 10.1080/17474086.2018.1447378] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Response to therapy is affected by the genetic heterogeneity of acute myeloid leukemia (AML) and persistence of leukemic cells below the threshold of morphological complete remission (mCR). Such persistence is called minimal (or measurable) residual disease (MRD). Areas covered: MRD assessment allows early identification of patients who are at high risk of relapse and who should timely receive aggressive therapy (e.g. allogeneic stem cell transplantation) and of those with a good quality mCR in whom an aggressive front-line therapy can be spared, avoiding the harm of excessive treatment toxicity. The most exploited methods to assess MRD are multiparameter flow cytometry (via identification of immunophenotypic aberrancies) or PCR-based assays (via identification of cytogenetic/molecular abnormalities). Expert commentary: A growing body of evidences demonstrates that positive MRD-testing at various time-points throughout the treatment course identifies patients at high risk of relapse. We will focus on the role of MRD as a biomarker to refine risk assessment and to prospectively direct treatment choices in adult with AML.
Collapse
Affiliation(s)
- Francesco Buccisano
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Luca Maurillo
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | | | - Ambra Di Veroli
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Eleonora De Bellis
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Annalisa Biagi
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Annagiulia Zizzari
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Valentina Rossi
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Vito Rapisarda
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Sergio Amadori
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Maria Teresa Voso
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Francesco Lo-Coco
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - William Arcese
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| | - Adriano Venditti
- a Department of Biomedicine and Prevention , University Tor Vergata of Rome , Rome , Italy
| |
Collapse
|
213
|
Gomez-Arteaga A, Guzman ML. Minimal Residual Disease in Acute Myeloid Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1100:111-125. [PMID: 30411263 DOI: 10.1007/978-3-319-97746-1_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Monitoring measurable (minimal) residual disease (MRD) in acute myeloid leukemia (AML) has greatly increased our ability to assess chemosensisitivity to treatment as well as the duration of treatment responses. There is strong evidence to support its prognostic value for long-term outcomes at different time points and across assays and targets. It's role as a surrogate endpoint to define risk-adapted strategies is still under evaluation. In this chapter, we will discuss the definition of MRD in AML, the potential contribution of leukemia stem cells (LSCs) to MRD and we will review all the current approaches to assess residual disease including the 2018 European Leukemia Network (ELN) working group recommendations for MRD standardization in AML. In addition, a summary of MRD studies associated to prognosis will be described.
Collapse
Affiliation(s)
- Alexandra Gomez-Arteaga
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine - New York Presbyterian Hospital, New York, NY, USA
| | - Monica L Guzman
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine - New York Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
214
|
Chang YJ, Zhao XS, Wang Y, Liu YR, Xu LP, Zhang XH, Chen H, Chen YH, Han W, Sun YQ, Yan CH, Mo XD, Liu KY, Huang XJ. Effects of pre- and post-transplantation minimal residual disease on outcomes in pediatric patients with acute myeloid leukemia receiving human leukocyte antigen-matched or mismatched related donor allografts. Am J Hematol 2017; 92:E659-E661. [PMID: 28929514 DOI: 10.1002/ajh.24910] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 01/25/2023]
Affiliation(s)
- Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
- Collaborative Innovation Center of Hematology, Peking University; Haidian Qu Beijing Shi China
| | - Xiao-Su Zhao
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
- Collaborative Innovation Center of Hematology, Peking University; Haidian Qu Beijing Shi China
| | - Yu Wang
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
- Collaborative Innovation Center of Hematology, Peking University; Haidian Qu Beijing Shi China
| | - Yan-Rong Liu
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Lan-Ping Xu
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Xiao-Hui Zhang
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
- Collaborative Innovation Center of Hematology, Peking University; Haidian Qu Beijing Shi China
| | - Huan Chen
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Yu-Hong Chen
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Wei- Han
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Yu-Qian Sun
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Chen-Hua Yan
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Xiao-Dong Mo
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Kai-Yan Liu
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Beijing 100044 P.R.C
- Collaborative Innovation Center of Hematology, Peking University; Haidian Qu Beijing Shi China
- Peking-Tsinghua Center for Life Sciences; Beijing 100871 China
| |
Collapse
|
215
|
Candoni A, De Marchi F, Zannier ME, Lazzarotto D, Filì C, Dubbini MV, Rabassi N, Toffoletti E, Lau BW, Fanin R. High prognostic value of pre-allogeneic stem cell transplantation minimal residual disease detection by WT1 gene expression in AML transplanted in cytologic complete remission. Leuk Res 2017; 63:22-27. [PMID: 29096332 DOI: 10.1016/j.leukres.2017.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 11/17/2022]
Abstract
We analyzed the outcome of allogeneic stem cell transplantation (allo-SCT) in acute myeloid leukemia (AML) patients according to molecular Minimal Residual Disease (MRD) status prior to allo-SCT. MRD was assessed by the quantitative expression of the pan-leukemic marker Wilms' tumor (WT1) gene, according to the validated LeukemiaNet method. Between 2005 and 2016, 122 consecutive AML patients, WT1 positive at diagnosis, received allo-SCT in cytologic complete remission (cCR). The median age at SCT was 53 years (range 18-70). Quantitative analysis of WT1 gene expression (bone marrow samples) was available in all cases both at diagnosis (100% of samples overexpressed WT1 with a mean of 8607±8187 copies/104 Abelson) and immediately before allo-SCT. Eighty one cases (66%) were MRD-WT1 negative (WT1 <250 copies) and 41/122 (44%) cases were MRD-WT1 positive (WT1 >250 copies) prior to allo-SCT. We evaluated post-SCT overall survival (OS), disease free survival (DFS) and relapse rate (RR), according to MRD-WT1 status pre-SCT. Both post-allo-SCT OS and DFS were significantly improved in patients who were MRD-WT1 negative at the time of SCT compared with those who were MRD-WT1 positive, with a median OS and DFS not reached in the MRD-WT1 negative group and 9 and 8 months, respectively, in the WT1 positive group (OS log-rank p<0.0001; hazard ratio [HR] 3.9, 95% confidence interval [95% CI] 2.0-7.38; DFS log-rank p<0.0001; HR 3.73, 95% CI 2.0-6.72). The RR after SCT was 15% (12/81) in pre-SCT MRD-WT1 negative cases and 44% (18/41) in MRD-WT1 positive cases (p=0.00073). Univariate analysis showed that MRD-WT1 negativity pre-SCT and grade <2 acute GVHD were significant prognostic factors for improved OS and DFS. However multivariate analysis showed MRD-WT1 negativity pre-SCT was the only independent prognostic factor for improved OS and DFS. These data show that pre allo-SCT molecular MRD evaluation using WT1 expression is a powerful predictor of post allo-SCT outcomes in AML undergoing SCT in cCR. Patients with both cCR and MRD-WT1 negativity before SCT have a very good outcome with lower RR and improved OS. The pre allo-SCT MRD-WT1 stratification in AML is a valuable tool to identify patients at high risk of post-SCT relapse, and can influence conditioning regimen intensification and/or post-SCT preemptive strategies.
Collapse
Affiliation(s)
- Anna Candoni
- Division of Hematology and SCT, University Hospital, Udine, Udine, Italy.
| | - Federico De Marchi
- Division of Hematology and SCT, University Hospital, Udine, Udine, Italy
| | | | - Davide Lazzarotto
- Division of Hematology and SCT, University Hospital, Udine, Udine, Italy
| | - Carla Filì
- Division of Hematology and SCT, University Hospital, Udine, Udine, Italy
| | | | - Nicholas Rabassi
- Division of Hematology and SCT, University Hospital, Udine, Udine, Italy
| | | | - Bonnie W Lau
- Department of Pediatric Hematology/Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Renato Fanin
- Division of Hematology and SCT, University Hospital, Udine, Udine, Italy
| |
Collapse
|
216
|
Importance of conditioning regimen intensity, MRD positivity, and KIR ligand mismatch in UCB transplantation. Bone Marrow Transplant 2017; 53:97-100. [PMID: 28991246 DOI: 10.1038/bmt.2017.212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
217
|
Jentzsch M, Bill M, Grimm J, Schulz J, Goldmann K, Beinicke S, Häntschel J, Pönisch W, Franke GN, Vucinic V, Behre G, Lange T, Niederwieser D, Schwind S. High BAALC copy numbers in peripheral blood prior to allogeneic transplantation predict early relapse in acute myeloid leukemia patients. Oncotarget 2017; 8:87944-87954. [PMID: 29152132 PMCID: PMC5675684 DOI: 10.18632/oncotarget.21322] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
High BAALC expression levels at acute myeloid leukemia diagnosis have been linked to adverse outcomes. Recent data indicate that high BAALC expression levels may also be used as marker for residual disease following acute myeloid leukemia treatment. Allogeneic hematopoietic stem cell transplantation (HSCT) offers a curative treatment for acute myeloid leukemia patients. However, disease recurrence remains a major clinical challenge and identification of high-risk patients prior to HSCT is crucial to improve outcomes. We performed absolute quantification of BAALC copy numbers in peripheral blood prior (median 7 days) to HSCT in complete remission (CR) or CR with incomplete peripheral recovery in 82 acute myeloid leukemia patients using digital droplet PCR (ddPCR) technology. An optimal cut-off of 0.14 BAALC/ABL1 copy numbers was determined and applied to define patients with high or low BAALC/ABL1 copy numbers. High pre-HSCT BAALC/ABL1 copy numbers significantly associated with higher cumulative incidence of relapse and shorter overall survival in univariable and multivariable models. Patients with high pre-HSCT BAALC/ABL1 copy numbers were more likely to experience relapse within 100 days after HSCT. Evaluation of pre-HSCT BAALC/ABL1 copy numbers in peripheral blood by ddPCR represents a feasible and rapid way to identify acute myeloid leukemia patients at high risk of early relapse after HSCT. The prognostic impact was also observed independently of other known clinical, genetic, and molecular prognosticators. In the future, prospective studies should evaluate whether acute myeloid leukemia patients with high pre-HSCT BAALC/ABL1 copy numbers benefit from additional treatment before or early intervention after HSCT.
Collapse
Affiliation(s)
- Madlen Jentzsch
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Marius Bill
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Juliane Grimm
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Julia Schulz
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Karoline Goldmann
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Stefanie Beinicke
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Janine Häntschel
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Wolfram Pönisch
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | | | - Vladan Vucinic
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Gerhard Behre
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Thoralf Lange
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | | | - Sebastian Schwind
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
218
|
Outcome and Minimal Residual Disease Monitoring in Patients with t(16;21) Acute Myelogenous Leukemia Undergoing Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2017; 24:163-168. [PMID: 28939454 DOI: 10.1016/j.bbmt.2017.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 09/01/2017] [Indexed: 01/01/2023]
Abstract
Patients with t(16;21) acute myelogenous leukemia (AML) who receive chemotherapy have poor outcomes. The treatment efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT) must be identified, and the usefulness of minimal residual disease (MRD) monitoring requires evaluation. Fourteen consecutive patients with t(16;21) AML undergoing allo-HSCT at our institution were included in this study. Translocation liposarcoma- ETS-related gene (TLS-ERG) transcript levels were serially monitored for a median of 15 months (range, 3-51 months) after allo-HSCT. Eight patients relapsed, 7 patients died from relapse-related causes, and 1 patient died from a non-relapse-related cause. The 2-year cumulative incidence rates of relapse, disease-free survival, and overall survival after HSCT were 66.2%, 30.8%, and 46.2%, respectively. Of the 3 patients who received an HLA-matched sibling transplant, 2 relapsed, and 1 (33.3%) was in hematologic complete remission (CR) but died of nonrelapse mortality, whereas 5 of 11 patients (45.5%) who received haploidentical transplantation were in CR and were alive. Two of 6 patients with undetectable TLS-ERG at the time of allo-HSCT relapsed, at 14 and 15 months, and 3 of 4 PCR-positive patients relapsed, at a median of 10 months after HSCT. Four patients with continually low post-HSCT TLS-ERG levels (mostly <.01%) remained alive and in CR. The TLS-ERG levels of all 8 patients who relapsed were significantly increased before the relapse, exceeding 1.0% in all 7 patients who experienced hematologic relapse. In total, 7 patients received modified donor lymphocyte infusion (DLI), and 1 patient received IFN-α. All 7 patients with a TLS-ERG level >5.0% at the time of intervention experienced an increase or a brief decrease in TLS-ERG level, followed by an increase, and 6 relapsed, whereas the TLS-ERG level of 1 patient with a TLS-ERG level <1.0% at intervention decreased to undetectable. Therefore, t(16;21) AML is an indication for allo-HSCT. Among the HSCT recipients, 30.8% responded to treatment with CR. TLS-ERG transcript levels reflect MRD and might predict relapse and guide effective intervention.
Collapse
|
219
|
Roloff GW, Lai C, Hourigan CS, Dillon LW. Technical Advances in the Measurement of Residual Disease in Acute Myeloid Leukemia. J Clin Med 2017; 6:jcm6090087. [PMID: 28925935 PMCID: PMC5615280 DOI: 10.3390/jcm6090087] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/09/2017] [Accepted: 09/13/2017] [Indexed: 12/31/2022] Open
Abstract
Outcomes for those diagnosed with acute myeloid leukemia (AML) remain poor. It has been widely established that persistent residual leukemic burden, often referred to as measurable or minimal residual disease (MRD), after induction therapy or at the time of hematopoietic stem cell transplant (HSCT) is highly predictive for adverse clinical outcomes and can be used to identify patients likely to experience clinically evident relapse. As a result of inherent genetic and molecular heterogeneity in AML, there is no uniform method or protocol for MRD measurement to encompass all cases. Several techniques focusing on identifying recurrent molecular and cytogenetic aberrations or leukemia-associated immunophenotypes have been described, each with their own strengths and weaknesses. Modern technologies enabling the digital quantification and tracking of individual DNA or RNA molecules, next-generation sequencing (NGS) platforms, and high-resolution imaging capabilities are among several new avenues under development to supplement or replace the current standard of flow cytometry. In this review, we outline emerging modalities positioned to enhance MRD detection and discuss factors surrounding their integration into clinical practice.
Collapse
Affiliation(s)
- Gregory W Roloff
- Myeloid Malignances Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Catherine Lai
- Myeloid Malignances Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Christopher S Hourigan
- Myeloid Malignances Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Laura W Dillon
- Myeloid Malignances Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
220
|
Haplo, we have a problem. Blood 2017; 130:1180. [PMID: 28882835 DOI: 10.1182/blood-2017-07-795062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
221
|
Is there a need for morphologic exam to detect relapse in AML if multi-parameter flow cytometry is employed? Leukemia 2017; 31:2536-2537. [DOI: 10.1038/leu.2017.263] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
222
|
Wall SA, Devine S, Vasu S. The who, how and why: Allogeneic transplant for acute myeloid leukemia in patients older than 60years. Blood Rev 2017; 31:362-369. [PMID: 28802907 DOI: 10.1016/j.blre.2017.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 05/26/2017] [Accepted: 07/11/2017] [Indexed: 10/19/2022]
Abstract
Acute myelogenous leukemia (AML) is primarily a disease of the elderly, and as such, our approach to treatment needs to be tailored to address an aging population. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains the only potentially curative treatment for intermediate and high risk AML, and until recently, its use had been limited to a younger population and dependent on availability of a donor. Advances in conditioning regimens, supportive care, and the use of alternative donor sources have greatly expanded access to this therapy. In this review, we summarize the challenges and unique biological aspects of treatment with allogeneic stem cell transplantation in this group of patients older than 60years. We also highlight areas of ongoing research including measurement of residual disease prior to and following transplant, post-remission maintenance therapy, and natural killer cell immunotherapy. Finally, we propose future directions for AML treatment in an elderly and aging population.
Collapse
Affiliation(s)
- Sarah A Wall
- Division of Hematology, Ohio State University, Columbus, OH, USA
| | - Steven Devine
- Division of Hematology, Ohio State University, Columbus, OH, USA
| | - Sumithira Vasu
- Division of Hematology, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
223
|
Hourigan CS, Gale RP, Gormley NJ, Ossenkoppele GJ, Walter RB. Measurable residual disease testing in acute myeloid leukaemia. Leukemia 2017; 31:1482-1490. [PMID: 28386105 DOI: 10.1038/leu.2017.113] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 12/24/2022]
Abstract
There is considerable interest in developing techniques to detect and/or quantify remaining leukaemia cells termed measurable or, less precisely, minimal residual disease (MRD) in persons with acute myeloid leukaemia (AML) in complete remission defined by cytomorphological criteria. An important reason for AML MRD-testing is the possibility of estimating the likelihood (and timing) of leukaemia relapse. A perfect MRD-test would precisely quantify leukaemia cells biologically able and likely to cause leukaemia relapse within a defined interval. AML is genetically diverse and there is currently no uniform approach to detecting such cells. Several technologies focused on immune phenotype or cytogenetic and/or molecular abnormalities have been developed, each with advantages and disadvantages. Many studies report a positive MRD-test at diverse time points during AML therapy identifies persons with a higher risk of leukaemia relapse compared with those with a negative MRD-test even after adjusting for other prognostic and predictive variables. No MRD-test in AML has perfect sensitivity and specificity for relapse prediction at the cohort- or subject levels and there are substantial rates of false-positive and -negative tests. Despite these limitations, correlations between MRD-test results and relapse risk have generated interest in MRD-test result-directed therapy interventions. However, convincing proof that a specific intervention will reduce relapse risk in persons with a positive MRD-test is lacking and needs testing in randomized trials. Routine clinical use of MRD-testing requires further refinements and standardization/harmonization of assay platforms and results reporting. Such data are needed to determine whether results of MRD-testing can be used as a surrogate end point in AML therapy trials. This could make drug-testing more efficient and accelerate regulatory approvals. Although MRD-testing in AML has advanced substantially, much remains to be done.
Collapse
Affiliation(s)
- C S Hourigan
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - R P Gale
- Haematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, UK
| | - N J Gormley
- Division of Hematology Products, Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - G J Ossenkoppele
- Division of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - R B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.,Department of Epidemiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
224
|
Percival ME, Lai C, Estey E, Hourigan CS. Bone marrow evaluation for diagnosis and monitoring of acute myeloid leukemia. Blood Rev 2017; 31:185-192. [PMID: 28190619 DOI: 10.1016/j.blre.2017.01.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/31/2017] [Indexed: 12/20/2022]
Abstract
The diagnosis of acute myeloid leukemia (AML) can be made based on peripheral blood or bone marrow blasts. In this review, we will discuss the role of bone marrow evaluation and peripheral blood monitoring in the diagnosis, management, and follow up of AML patients. For patients with circulating blasts, it is reasonable to perform the necessary studies needed for diagnosis and risk stratification, including multiparametric flow cytometry, cytogenetics, and molecular analysis, on a peripheral blood specimen. The day 14 marrow is used to document hypocellularity in response to induction chemotherapy, but it is unclear if that assessment is necessary as it often does not affect immediate management. Currently, response assessments performed at count recovery for evaluation of remission and measurable residual disease rely on bone marrow sampling. For monitoring of relapse, peripheral blood evaluation may be adequate, but the sensitivity of bone marrow testing is in some cases superior. While bone marrow evaluation can certainly be avoided in particular situations, this cumbersome and uncomfortable procedure currently remains the de facto standard for response assessment.
Collapse
Affiliation(s)
- Mary-Elizabeth Percival
- Department of Medicine, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Catherine Lai
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elihu Estey
- Department of Medicine, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Christopher S Hourigan
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|