101
|
Blätte TJ, Schmalbrock LK, Skambraks S, Lux S, Cocciardi S, Dolnik A, Döhner H, Döhner K, Bullinger L. getITD for FLT3-ITD-based MRD monitoring in AML. Leukemia 2019; 33:2535-2539. [PMID: 31089248 PMCID: PMC8075860 DOI: 10.1038/s41375-019-0483-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Tamara J Blätte
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Laura K Schmalbrock
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Sabrina Skambraks
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Susanne Lux
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Sibylle Cocciardi
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Anna Dolnik
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany.
| |
Collapse
|
102
|
Molecular Detection of Minimal Residual Disease before Allogeneic Stem Cell Transplantation Predicts a High Incidence of Early Relapse in Adult Patients with NPM1 Positive Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:cancers11101455. [PMID: 31569375 PMCID: PMC6826431 DOI: 10.3390/cancers11101455] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 01/07/2023] Open
Abstract
We analyzed the impact of alloHSCT in a single center cohort of 89 newly diagnosed NPM1mut AML patients, consecutively treated according to the Northern Italy Leukemia Group protocol 02/06 [NCT00495287]. After two consolidation cycles, the detection of measurable residual disease (MRD) by RQ-PCR was strongly associated with an inferior three-year overall survival (OS, 45% versus 84%, p = 0.001) and disease-free survival (DFS, 44% versus 76%, p = 0.006). In MRD-negative patients, post-remissional consolidation with alloHSCT did not provide a significant additional benefit over a conventional chemotherapy in terms of overall survival [OS, 89% (95% CI 71–100%) versus 81% (95% CI 64–100%), p = 0.59] and disease-free survival [DFS, 80% (95% CI 59–100%) versus 75% (95% CI 56–99%), p = 0.87]. On the contrary, in patients with persistent MRD positivity, the three-year OS and DFS were improved in patients receiving an alloHSCT compared to those allocated to conventional chemotherapy (OS, 52% versus 31%, p = 0.45 and DFS, 50% versus 17%, p = 0.31, respectively). However, in this group of patients, the benefit of alloHSCT was still hampered by a high incidence of leukemia relapse during the first year after transplantation (43%, 95% CI 25–60%). Consolidative alloHSCT improves outcomes compared to standard chemotherapy in patients with persistent NPM1mut MRD positivity, but in these high-risk patients, the significant incidence of leukemia relapse must be tackled by post-transplant preemptive treatments.
Collapse
|
103
|
FERMI: A Novel Method for Sensitive Detection of Rare Mutations in Somatic Tissue. G3-GENES GENOMES GENETICS 2019; 9:2977-2987. [PMID: 31352405 PMCID: PMC6723130 DOI: 10.1534/g3.119.400438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
With growing interest in monitoring mutational processes in normal tissues, tumor heterogeneity, and cancer evolution under therapy, the ability to accurately and economically detect ultra-rare mutations is becoming increasingly important. However, this capability has often been compromised by significant sequencing, PCR and DNA preparation error rates. Here, we describe FERMI (Fast Extremely Rare Mutation Identification) - a novel method designed to eliminate the majority of these sequencing and library-preparation errors in order to significantly improve rare somatic mutation detection. This method leverages barcoded targeting probes to capture and sequence DNA of interest with single copy resolution. The variant calls from the barcoded sequencing data are then further filtered in a position-dependent fashion against an adaptive, context-aware null model in order to distinguish true variants. As a proof of principle, we employ FERMI to probe bone marrow biopsies from leukemia patients, and show that rare mutations and clonal evolution can be tracked throughout cancer treatment, including during historically intractable periods like minimum residual disease. Importantly, FERMI is able to accurately detect nascent clonal expansions within leukemias in a manner that may facilitate the early detection and characterization of cancer relapse.
Collapse
|
104
|
Agrawal M, Schwarz P, Giaimo BD, Bedzhov I, Corbacioglu A, Weber D, Gaidzik VI, Jahn N, Rücker FG, Schroeder T, Kindler T, Wattad M, Götze K, Lübbert M, Salwender H, Ringhoffer M, Lange E, Koller E, Thol F, Heuser M, Ganser A, Bullinger L, Paschka P, Döhner H, Geiger H, Borggrefe T, Döhner K, Oswald F. Functional and clinical characterization of the alternatively spliced isoform AML1-ETO9a in adult patients with translocation t(8;21)(q22;q22.1) acute myeloid leukemia (AML). Leukemia 2019; 34:630-634. [PMID: 31462736 PMCID: PMC7214266 DOI: 10.1038/s41375-019-0551-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/29/2019] [Accepted: 06/06/2019] [Indexed: 12/04/2022]
Affiliation(s)
- Mridul Agrawal
- Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany
| | - Peggy Schwarz
- Klinik für Innere Medizin I, Universitätsklinikum Ulm, Ulm, Germany
| | | | - Ivan Bedzhov
- Embryonic Self-Organization research group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149, Münster, Germany
| | | | - Daniela Weber
- Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany
| | - Verena I Gaidzik
- Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany
| | - Nikolaus Jahn
- Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany
| | - Frank G Rücker
- Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany
| | - Thomas Schroeder
- Klinik für Hämatologie, Onkologie und Klinische Immunologie, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Thomas Kindler
- III. Medizinische Klinik und Poliklinik, Universitätsmedizin Mainz, Mainz, Germany
| | - Mohammed Wattad
- Klinik für Hämatologie, Internistische Onkologie und Stammzellentransplantation, Evangelisches Krankenhaus Essen-Werden, Essen, Germany
| | - Katharina Götze
- III. Medizinische Klinik, Klinikum rechts der Isar der Technischen Universität München, München, Germany
| | - Michael Lübbert
- Klinik für Innere Medizin I, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Hans Salwender
- II. Medizinische Abteilung, Asklepios Klinik Altona, Hamburg, Germany
| | - Mark Ringhoffer
- Medizinische Klinik III, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - Elisabeth Lange
- Klinik für Hämatologie, Onkologie und Palliativmedizin, Evangelisches Krankenhaus Hamm, Hamm, Germany
| | - Elisabeth Koller
- Medizinische Abteilung, Hanusch-Krankenhaus der WGKK, Wien, Austria
| | - Felicitas Thol
- Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Medizinische Hochschule Hannover, Hannover, Germany
| | - Michael Heuser
- Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Medizinische Hochschule Hannover, Hannover, Germany
| | - Arnold Ganser
- Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Medizinische Hochschule Hannover, Hannover, Germany
| | - Lars Bullinger
- Medizinische Klinik m. S. Hämatologie, Onkologie und Tumorimmunologie, Campus Virchow-Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Paschka
- Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany
| | - Hartmut Döhner
- Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany
| | - Hartmut Geiger
- Institut für Molekulare Medizin, Universität Ulm, Ulm, Germany.,Division of Experimental Hematology and Cancer Biology, CCHMC, Cincinnati, OH, USA
| | - Tilman Borggrefe
- Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Konstanze Döhner
- Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany.
| | - Franz Oswald
- Klinik für Innere Medizin I, Universitätsklinikum Ulm, Ulm, Germany
| |
Collapse
|
105
|
Short NJ, Ravandi F. How close are we to incorporating measurable residual disease into clinical practice for acute myeloid leukemia? Haematologica 2019; 104:1532-1541. [PMID: 31273094 PMCID: PMC6669140 DOI: 10.3324/haematol.2018.208454] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/05/2019] [Indexed: 12/22/2022] Open
Abstract
Assessment of measurable residual disease, also called "minimal residual disease," in patients with acute myeloid leukemia in morphological remission provides powerful prognostic information and complements pretreatment factors such as cytogenetics and genomic alterations. Based on data that low levels of persistent or recurrent residual leukemia are consistently associated with an increased risk of relapse and worse long-term outcomes, its routine assessment has been recommended by some experts and consensus guidelines. In addition to providing important prognostic information, the detection of measurable residual disease may also theoretically help to determine the optimal post-remission strategy for an individual patient. However, the full therapeutic implications of measurable residual disease are uncertain and thus controversy exists as to whether it should be routinely incorporated into clinical practice. While some evidence supports the use of allogeneic stem cell transplantation or hypomethylating agents for some subgroups of patients in morphological remission but with detectable residual leukemia, the appropriate use of this information in making clinical decisions remains largely speculative at present. To resolve this pressing clinical issue, several ongoing studies are evaluating measurable residual disease-directed treatments in acute myeloid leukemia and may lead to new, effective strategies for patients in these circumstances. This review examines the common technologies used in clinical practice and in the research setting to detect residual leukemia, the major clinical studies establishing the prognostic impact of measurable residual disease in acute myeloid leukemia, and the potential ways, both now and in the future, that such testing may rationally guide therapeutic decision-making.
Collapse
Affiliation(s)
- Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
106
|
Guolo F, Minetto P, Clavio M, Miglino M, Colombo N, Cagnetta A, Cea M, Marcolin R, Todiere A, Ballerini F, Gobbi M, Lemoli RM. Longitudinal minimal residual disease (MRD) evaluation in acute myeloid leukaemia with
NPM1
mutation: from definition of molecular relapse to MRD‐driven salvage approach. Br J Haematol 2019; 186:e223-e225. [DOI: 10.1111/bjh.16116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Fabio Guolo
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Paola Minetto
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Marino Clavio
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Maurizio Miglino
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Nicoletta Colombo
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Antonia Cagnetta
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Michele Cea
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Riccardo Marcolin
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Andrea Todiere
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Filippo Ballerini
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Marco Gobbi
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Roberto Massimo Lemoli
- Clinic of Haematology, Department of Internal Medicine (DiMI) University of Genoa Genoa Italy
- IRCCS Ospedale Policlinico San Martino Genoa Italy
| |
Collapse
|
107
|
|
108
|
Zhang Y, Zhang YM, Zhang YS, Tang GS, Zhang WP, Yang JM, Wang JM, Hu XX. [Prognostic significance of minimal residual disease before post-remission therapy in younger adult acute myeloid leukemia patients with intermediate risk and negative of FLT3-ITD, NPM1 and biallelic CEBPA mutations]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:597-601. [PMID: 32397025 PMCID: PMC7364900 DOI: 10.3760/cma.j.issn.0253-2727.2019.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Y Zhang
- Department of Hematology, Changhai Hospital, the Second Military Medical University; Institute of Hematologic Disease of Chinese PLA, Shanghai 200433, China
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Ossenkoppele G, Schuurhuis GJ, van de Loosdrecht A, Cloos J. Can we incorporate MRD assessment into clinical practice in AML? Best Pract Res Clin Haematol 2019; 32:186-191. [DOI: 10.1016/j.beha.2019.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/08/2019] [Indexed: 12/29/2022]
|
110
|
Abstract
Increasing evidence supports the prognostic significance of measurable residual disease (MRD) in acute myeloid leukemia (AML). Dynamic MRD assessment for patients with AML complements baseline patient risk assessment factors in determining patient prognosis. MRD status may also be helpful in informing therapeutic decisions. The European Leukemia Net MRD working party recently issued consensus recommendations for the use of MRD in AML. The Food and Drug Administration also issued advice for using MRD in trials of hematologic malignancies. This article discusses MRD testing, highlights the challenges in adopting MRD testing in clinical practice, and provides insights into the future of the field.
Collapse
|
111
|
Heuser M, Mina A, Stein EM, Altman JK. How Precision Medicine Is Changing Acute Myeloid Leukemia Therapy. Am Soc Clin Oncol Educ Book 2019; 39:411-420. [PMID: 31099617 DOI: 10.1200/edbk_238687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pretreatment somatic mutations influence acute myeloid leukemia (AML) pathogenesis and responses to chemotherapy. Integration of cytogenetic abnormalities and molecular mutations, co-occurring and in isolation, have resulted in a more refined prognostic assessment. In addition, research performed over the last few years has led to the development of novel therapies and new drug approvals in patients with both newly diagnosed and relapsed/refractory (R/R) AML. Here we discuss the use of these newly approved therapies. Advances in AML have also occurred through development of better tools to assess response to treatment. Both multiparameter flow cytometry and polymerase chain reaction can be used to assess for the presence or absence of measurable residual disease (MRD) and increase the sensitivity of response assessment. The role of MRD assessment is gaining relevance and its integration in clinical trials and treatment decision making will be explored in the second half of this article.
Collapse
Affiliation(s)
| | | | - Eytan M Stein
- 3 Memorial Sloan-Kettering Cancer Center and Weil Cornell Medical College, New York, NY
| | | |
Collapse
|
112
|
Cocciardi S, Dolnik A, Kapp-Schwoerer S, Rücker FG, Lux S, Blätte TJ, Skambraks S, Krönke J, Heidel FH, Schnöder TM, Corbacioglu A, Gaidzik VI, Paschka P, Teleanu V, Göhring G, Thol F, Heuser M, Ganser A, Weber D, Sträng E, Kestler HA, Döhner H, Bullinger L, Döhner K. Clonal evolution patterns in acute myeloid leukemia with NPM1 mutation. Nat Commun 2019; 10:2031. [PMID: 31048683 PMCID: PMC6497712 DOI: 10.1038/s41467-019-09745-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 03/28/2019] [Indexed: 12/15/2022] Open
Abstract
Mutations in the nucleophosmin 1 (NPM1) gene are considered founder mutations in the pathogenesis of acute myeloid leukemia (AML). To characterize the genetic composition of NPM1 mutated (NPM1mut) AML, we assess mutation status of five recurrently mutated oncogenes in 129 paired NPM1mut samples obtained at diagnosis and relapse. We find a substantial shift in the genetic pattern from diagnosis to relapse including NPM1mut loss (n = 11). To better understand these NPM1mut loss cases, we perform whole exome sequencing (WES) and RNA-Seq. At the time of relapse, NPM1mut loss patients (pts) feature distinct mutational patterns that share almost no somatic mutation with the corresponding diagnosis sample and impact different signaling pathways. In contrast, profiles of pts with persistent NPM1mut are reflected by a high overlap of mutations between diagnosis and relapse. Our findings confirm that relapse often originates from persistent leukemic clones, though NPM1mut loss cases suggest a second "de novo" or treatment-associated AML (tAML) as alternative cause of relapse.
Collapse
Affiliation(s)
- Sibylle Cocciardi
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Anna Dolnik
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Silke Kapp-Schwoerer
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Frank G Rücker
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Susanne Lux
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Tamara J Blätte
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Sabrina Skambraks
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Jan Krönke
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Florian H Heidel
- Department of Internal Medicine II, Hematology and Oncology, Friedrich-Schiller-University Medical Center, Jena, 07743, Germany.,Leibniz-Institute on Aging, Fritz-Lipmann-Institute, Jena, 07745, Germany
| | - Tina M Schnöder
- Department of Internal Medicine II, Hematology and Oncology, Friedrich-Schiller-University Medical Center, Jena, 07743, Germany.,Leibniz-Institute on Aging, Fritz-Lipmann-Institute, Jena, 07745, Germany
| | - Andrea Corbacioglu
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Verena I Gaidzik
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Peter Paschka
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Veronica Teleanu
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Gudrun Göhring
- Institute of Cell & Molecular Pathology, Hannover Medical School, Hannover, 30625, Germany
| | - Felicitas Thol
- Department of Haematology, Haemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, 30625, Germany
| | - Michael Heuser
- Department of Haematology, Haemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, 30625, Germany
| | - Arnold Ganser
- Department of Haematology, Haemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, 30625, Germany
| | - Daniela Weber
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Eric Sträng
- Institute of Medical Systems Biology, Ulm University, Ulm, 30625, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, 30625, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Lars Bullinger
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany. .,Department of Hematology, Oncology and Tumorimmunology, Charité University Medicine, Berlin, 13353, Germany.
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany.
| |
Collapse
|
113
|
Heiblig M, Labussière-Wallet H, Nicolini FE, Michallet M, Hayette S, Sujobert P, Plesa A, Balsat M, Paubelle E, Barraco F, Tigaud I, Ducastelle S, Wattel E, Salles G, Thomas X. Prognostic Value of Genetic Alterations in Elderly Patients with Acute Myeloid Leukemia: A Single Institution Experience. Cancers (Basel) 2019; 11:cancers11040570. [PMID: 31013658 PMCID: PMC6520979 DOI: 10.3390/cancers11040570] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/23/2019] [Accepted: 04/08/2019] [Indexed: 12/15/2022] Open
Abstract
Although the outcome in younger adults with acute myeloid leukemia (AML) has improved, the benefit associated with standard intensive chemotherapy in older patients remains debatable. In this study, we investigated the incidence and the prognostic significance of genetic characteristics according to treatment intensity in patients aged 60 years or older. On the 495 patients of our cohort, DNMT3AR882 (25.2%), NPM1 (23.7%) and FLT3-ITD (16.8%) were the most frequent molecular mutations found at diagnosis. In this elderly population, intensive chemotherapy seemed to be a suitable option in terms of early death and survival, except for normal karyotype (NK) NPM1−FLT3-ITD+ patients and those aged over 70 within the adverse cytogenetic/molecular risk group. The FLT3-ITD mutation was systematically associated with an unfavorable outcome, independently of the ratio. NK NPM1+/FLT3-TKD+ genotype tends to confer a good prognosis in patients treated intensively. Regarding minimal residual disease prognostic value, overall survival was significantly better for patients achieving a 4 log NPM1 reduction (median OS: 24.4 vs. 12.8 months, p = 0.013) but did not reach statistical significance for progression free survival. This retrospective study highlights that intensive chemotherapy may not be the most appropriate option for each elderly patient and that molecular markers may help treatment intensity decision-making.
Collapse
Affiliation(s)
- Maël Heiblig
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
- University Claude Bernard Lyon 1, 69100 Villeurbanne, France.
| | - Hélène Labussière-Wallet
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | | | - Mauricette Michallet
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Sandrine Hayette
- Laboratory of Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Pierre Sujobert
- Laboratory of Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Adriana Plesa
- Laboratory of Cytology and Immunology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Marie Balsat
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Etienne Paubelle
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Fiorenza Barraco
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Isabelle Tigaud
- Laboratory of Cytogenetics, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Sophie Ducastelle
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Eric Wattel
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Gilles Salles
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| | - Xavier Thomas
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France.
| |
Collapse
|
114
|
Salehzadeh S, Guerrini F, Pizzano U, Grassi S, Ciabatti E, Iovino L, Buda G, Caracciolo F, Benedetti E, Orciuolo E, Pelosini M, Consani G, Carulli G, Metelli MR, Martini F, Mazziotta F, Mazzantini E, Rossi P, Tavarozzi R, Ricci F, Petrini M, Galimberti S. The assessment of minimal residual disease versus that of somatic mutations for predicting the outcome of acute myeloid leukemia patients. Cancer Cell Int 2019; 19:83. [PMID: 30992690 PMCID: PMC6449954 DOI: 10.1186/s12935-019-0807-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/28/2019] [Indexed: 12/22/2022] Open
Abstract
Background In addition to morphological and cytogenetic features, acute myeloid leukemias are characterized by mutations that can be used for target-therapy; also the minimal/measurable residual disease (MRD) could be an important prognostic factor. The purpose of this retrospective study was to investigate if somatic mutations could represent an additional prognostic value in respect of MRD alone. Method At baseline, 98 patients were tested for NPM1, FLT3, and for WT1 expression; 31 for ASXL1, TET2, IDH1, IDH2, N-RAS, WT1, c-KIT, RUNX1, and DNMT3A. The same genes have been also tested after induction and consolidation. Results Overall, 60.2% of our patients resulted mutated: 24.5% carried mutations of FLT3-ITD, 38.7% of NPM1, 48.4% of c-KIT, 25.8% of N-RAS and 19.3% of IDH2. The probability of achieving a complete response (CR) was higher for younger patients, with low ELN risk score, NPM1-mutated, with low WT1 levels, and without FLT3. The presence of additional mutations represented a poor predictive factor: only 19% of these cases achieved CR in comparison to 43% of subjects without any of it. Concerning survival, it was conditioned by a lower ELN risk score, younger age, reduction > 1 log of the NPM1 mutational burden, disappearance of FLT3 mutations and lower WT1 expression. Regarding the role of the additional mutations, they impaired the outcome of 20% of the already MRD-negative patients. Concerning the possibility of predicting relapse, we observed an increase of the NPM1 mutational burden at the time-point immediately preceding the relapse (about 2 months earlier) in 50% of subjects. Similarly concerning WT1, an increase of its expression anticipated disease recurrence in 64% of cases. Conclusions We demonstrated that additional somatic mutations are able to impair outcome of the already MRD-negative subjects. About MRD, we suggest a prognostic role also for the WT1 expression. Finally, we considered as relevant the assessment of NPM1 quantity clearance instead of the presence/absence of mutations alone. Still remains in doubt the utility in terms of long-term prognosis of a baseline more complex mutational screening; we could hypothesize that it would be useful for those patients where other markers are not available or who reached the MRD negativity. Electronic supplementary material The online version of this article (10.1186/s12935-019-0807-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Serena Salehzadeh
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy.,4University of Rome Tor Vergata, Rome, Italy.,5Ospedale S. Chiara, UO Ematologia, Via Roma, 67, 56126 Pisa, Italy
| | - Francesca Guerrini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Umberto Pizzano
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Susanna Grassi
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Elena Ciabatti
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Lorenzo Iovino
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Gabriele Buda
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Francesco Caracciolo
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Edoardo Benedetti
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Enrico Orciuolo
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Matteo Pelosini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Giovanni Consani
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Giovanni Carulli
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | | | - Francesca Martini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Francesco Mazziotta
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy.,2GeNOMEC School of Doctorate, University of Siena, Siena, Italy
| | - Elisa Mazzantini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Pietro Rossi
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Rita Tavarozzi
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Federica Ricci
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Mario Petrini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Sara Galimberti
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| |
Collapse
|
115
|
Versluis J, Cornelissen JJ. Risks and benefits in a personalized application of allogeneic transplantation in patients with AML in first CR. Semin Hematol 2019; 56:164-170. [DOI: 10.1053/j.seminhematol.2018.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/29/2018] [Accepted: 08/29/2018] [Indexed: 11/11/2022]
|
116
|
Levine RL, Valk PJM. Next-generation sequencing in the diagnosis and minimal residual disease assessment of acute myeloid leukemia. Haematologica 2019; 104:868-871. [PMID: 30923100 PMCID: PMC6518900 DOI: 10.3324/haematol.2018.205955] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/14/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA .,Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter J M Valk
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA .,Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
117
|
Ehinger M, Pettersson L. Measurable residual disease testing for personalized treatment of acute myeloid leukemia. APMIS 2019; 127:337-351. [PMID: 30919505 DOI: 10.1111/apm.12926] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022]
Abstract
This review summarizes - with the practicing hematologist in mind - the methods used to determine measurable residual disease (MRD) in everyday practice with some future perspectives, and the current knowledge about the prognostic impact of MRD on outcome in acute myeloid leukemia (AML), excluding acute promyelocytic leukemia. Possible implications for choice of MRD method, timing of MRD monitoring, and guidance of therapy are discussed in general and in some detail for certain types of leukemia with specific molecular markers to monitor, including core binding factor (CBF)-leukemias and NPM1-mutated leukemias.
Collapse
Affiliation(s)
- Mats Ehinger
- Department of Clinical Sciences, Pathology, Skane University Hospital, Lund University, Lund, Sweden
| | - Louise Pettersson
- Department of Pathology, Halland Hospital Halmstad, Region Halland, Halmstad, Sweden.,Faculty of Medicine, Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
118
|
Roug AS, Ommen HB. Clinical Use of Measurable Residual Disease in Acute Myeloid Leukemia. Curr Treat Options Oncol 2019; 20:28. [PMID: 30874904 DOI: 10.1007/s11864-019-0627-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OPINION STATEMENT Treatment of acute myeloid leukemia (AML) remains a high-risk venture for the patient suffering from the disease. There is a real risk of succumbing to the treatment rather than the disease, and even so, cure is much less than certain. Since the establishment of complete remission as a prerequisite for cure in the 1960s, a number of years passed before advanced techniques for detecting minute amounts of disease matured sufficiently for clinical implementation. The two main techniques for detection of measurable residual disease (MRD) remain qPCR and multicolor flow cytometry. When performed in expert laboratories, both these modalities offer treating physicians excellent opportunity to follow the amount of residual disease upon treatment and offer unparalleled prognostication. In some AML and age group subsets, evidence now exist to support the choice of both proceeding to allogeneic transplant and not doing so. In other AML subgroups, MRD has sufficient discriminative power to identify patients likely to benefit from allogeneic transplant and patients likely not to. After treatment or transplantation, follow-up by molecular techniques can, with high certainty, predict relapse months before bone marrow function deterioration. On the other hand, options upon so-called molecular relapse are less well tested but recent evidence supports the use of azacitidine both in transplanted patients and patients consolidated with chemotherapy. In conclusion, MRD testing during treatment is a superb prognosticator and a major tool when choosing whether a patient should be transplanted or not. The exact use of MRD testing after treatment is less well defined but evidence is mounting for the instigation of treatment upon rising MRD levels (pre-emptive treatment) before morphologically detectable relapse.
Collapse
Affiliation(s)
- Anne Stidsholt Roug
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, Mølleparkvej 4, DK-9000, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Hans Beier Ommen
- Department of Hematology, Aarhus University Hospital, Palle-Juul Jensens Boulevard 99, DK-8200, Aarhus N, Denmark.
| |
Collapse
|
119
|
A next-generation sequencing-based assay for minimal residual disease assessment in AML patients with FLT3-ITD mutations. Blood Adv 2019; 2:825-831. [PMID: 29643105 DOI: 10.1182/bloodadvances.2018015925] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/13/2018] [Indexed: 12/22/2022] Open
Abstract
Internal tandem duplications in fms-like tyrosine kinase 3 (FLT3-ITDs) are common in acute myeloid leukemia (AML) and confer a poor prognosis. A sensitive and specific assay for the detection of minimal residual disease (MRD) in FLT3-ITD mutated AML could guide therapy decisions. Existing assays for MRD in FLT3-ITD AML have not been particularly useful because of limited sensitivity. We developed a sensitive and specific MRD assay for FLT3-ITD mutations using next-generation sequencing. The initial validation of this assay was performed by spiking fixed amounts of mutant DNA into wild-type DNA to establish a sensitivity of detection equivalent to ≥1 FLT3-ITD-containing cell in 10 000, with a minimum input of 100 000 cell equivalents of DNA. We subsequently validated the assay in bone marrow samples from patients with FLT3-ITD AML in remission. Finally, we analyzed bone marrow samples from 80 patients with FLT3-ITD relapsed/refractory AML participating in a trial of a novel FLT3 inhibitor, gilteritinib, and demonstrated a relationship between the mutation burden, as detected by the assay, and overall survival. This novel MRD assay is specific and 2 orders of magnitude more sensitive than currently available polymerase chain reaction- or next-generation sequencing-based FLT3-ITD assays. The assay is being prospectively validated in ongoing randomized clinical trials.
Collapse
|
120
|
[Diagnosis and treatment of acute myeloid leukemia : The updated 2018 Onkopedia Guideline]. Internist (Berl) 2019; 60:257-272. [PMID: 30767029 DOI: 10.1007/s00108-019-0562-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In April 2018, an updated version of the previously published guidelines on acute myeloid leukemia (AML) from 2010 and 2017 was released. A revision was necessary because of two positive aspects: First, new data and insights on risk stratification and monitoring, and second, the clinical development and approval of new agents. The modified genetic risk classification allows a more precise distinction of different diagnostic groups and consequently a better matched post-remission treatment. The availability of new targeted drugs such as inhibitors turns genetic analyses from a mere prognostic tool into an instrument for treatment decisions. Several recently approved agents expand the treatment options for AML and raise hope for an improved prognosis and cure in the future.
Collapse
|
121
|
Next Generation Sequencing in AML-On the Way to Becoming a New Standard for Treatment Initiation and/or Modulation? Cancers (Basel) 2019; 11:cancers11020252. [PMID: 30795628 PMCID: PMC6406956 DOI: 10.3390/cancers11020252] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/08/2019] [Accepted: 02/12/2019] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disease caused by genetic abberations occurring predominantly in the elderly. Next generation sequencing (NGS) analysis has led to a deeper genetic understanding of the pathogenesis and the role of recently discovered genetic precursor lesions (clonal hematopoiesis of indeterminate/oncogenic potential (CHIP/CHOP)) in the evolution of AML. These advances are reflected by the inclusion of certain mutations in the updated World Health Organization (WHO) 2016 classification and current treatment guidelines by the European Leukemia Net (ELN) and National Comprehensive Cancer Network (NCCN) and results of mutational testing are already influencing the choice and timing of (targeted) treatment. Genetic profiling and stratification of patients into molecularly defined subgroups are expected to gain ever more weight in daily clinical practice. Our aim is to provide a concise summary of current evidence regarding the relevance of NGS for the diagnosis, risk stratification, treatment planning and response assessment in AML, including minimal residual disease (MRD) guided approaches. We also summarize recently approved drugs targeting genetically defined patient populations with risk adapted- and individualized treatment strategies.
Collapse
|
122
|
Deng DX, Zhu HH, Liu YR, Chang YJ, Ruan GR, Jia JS, Jiang H, Jiang Q, Zhao XS, Huang XJ. Minimal residual disease detected by multiparameter flow cytometry is complementary to genetics for risk stratification treatment in acute myeloid leukemia with biallelic CEBPA mutations. Leuk Lymphoma 2019; 60:2181-2189. [PMID: 30773106 DOI: 10.1080/10428194.2019.1576868] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Dao-Xing Deng
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Hong-Hu Zhu
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Yan-Rong Liu
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Ying-Jun Chang
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Guo-Rui Ruan
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Jin-Song Jia
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Hao Jiang
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Qian Jiang
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Xiao-Su Zhao
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Xiao-Jun Huang
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
123
|
Pasic I, Da'na W, Lam W, Law A, Lipton JH, Viswabandya A, Kim DD, Thyagu S, Messner HA, Michelis FV. Influence of FLT3-ITD and NPM1 status on allogeneic hematopoietic cell transplant outcomes in patients with cytogenetically normal AML. Eur J Haematol 2019; 102:368-374. [PMID: 30706524 DOI: 10.1111/ejh.13216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVE In individuals with cytogenetically normal (CN) AML, disease risk is estimated using molecular features such as the status of NPM1 and FLT3-ITD genes. However, data regarding the impact of NPM1 and FLT3-ITD status on hematopoietic stem cell transplant (HCT) outcomes are limited. We examined the effect of NPM1 and FLT3-ITD status on transplant outcomes in 131 CN AML patients transplanted at Princess Margaret Hospital between 2006 and 2017. METHODS Overall survival (OS) was calculated using Kaplan-Meier analysis and multivariable Cox proportional hazards regression. Cumulative incidence of relapse (CIR) and non-relapse mortality (NRM) were calculated using competing risk regression. RESULTS There was no difference in 3-year OS among NPM1+ /FLT3-ITD- , NPM1- /FLT3-ITD- , NPM1+ /FLT3-ITD+ and NPM1- /FLT3-ITD+ patients: 56% (95% CI, 29%-76%), 61% (95% CI, 46%-73%), 53% (95% CI, 34%-70%) and 52% (95% CI, 17%-78%), respectively. CIR at 3-years was similar among NPM1- /FLT3-ITD- , NPM1+ /FLT3-ITD+ and NPM1- /FLT3-ITD+ patients-14% (95% CI, 6%-26%), 13% (95% CI, 4%-28%) and 19% (95% CI, 4%-41%), respectively-while there were no relapses in the NPM1+ /FLT3-ITD- group. NRM at 3 years for NPM1+ /FLT3-ITD- , NPM1- /FLT3-ITD- , NPM1+ /FLT3-ITD+ and NPM1- /FLT3-ITD+ patients was similar at 44% (95% CI, 19%-67%), 38% (95% CI, 25%-50%), 43% (95% CI, 25%-59%) and 44% (95% CI, 14%-71%), respectively. CONCLUSION NPM1 and FLT3-ITD status may provide limited prognostic information about transplant outcomes in CN AML patients.
Collapse
Affiliation(s)
- Ivan Pasic
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Waleed Da'na
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Wilson Lam
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Arjun Law
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey H Lipton
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Auro Viswabandya
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Dennis D Kim
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Santhosh Thyagu
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hans A Messner
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Fotios V Michelis
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
124
|
Forghieri F, Riva G, Lagreca I, Barozzi P, Vallerini D, Morselli M, Paolini A, Bresciani P, Colaci E, Maccaferri M, Gilioli A, Nasillo V, Messerotti A, Pioli V, Arletti L, Giusti D, Bettelli F, Celli M, Donatelli F, Corradini G, Basso S, Gurrado A, Cellini M, Trenti T, Marasca R, Narni F, Martelli MP, Falini B, Potenza L, Luppi M, Comoli P. Characterization and dynamics of specific T cells against nucleophosmin-1 (NPM1)-mutated peptides in patients with NPM1-mutated acute myeloid leukemia. Oncotarget 2019; 10:869-882. [PMID: 30783516 PMCID: PMC6368236 DOI: 10.18632/oncotarget.26617] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/03/2019] [Indexed: 12/13/2022] Open
Abstract
Nucleophosmin(NPM1)-mutated protein, a leukemia-specific antigen, represents an ideal target for AML immunotherapy. We investigated the dynamics of NPM1-mutated-specific T cells on PB and BM samples, collected from 31 adult NPM1-mutated AML patients throughout the disease course, and stimulated with mixtures of 18 short and long peptides (9-18mers), deriving from the complete C-terminal of the NPM1-mutated protein. Two 9-mer peptides, namely LAVEEVSLR and AVEEVSLRK (13.9-14.9), were identified as the most immunogenic epitopes. IFNγ-producing NPM1-mutated-specific T cells were observed by ELISPOT assay after stimulation with peptides 13.9-14.9 in 43/85 (50.6%) PB and 34/80 (42.5%) BM samples. An inverse correlation between MRD kinetics and anti-leukemic specific T cells was observed. Cytokine Secretion Assays allowed to predominantly and respectively identify Effector Memory and Central Memory T cells among IFNγ-producing and IL2-producing T cells. Moreover, NPM1-mutated-specific CTLs against primary leukemic blasts or PHA-blasts pulsed with different peptide pools could be expanded ex vivo from NPM1-mutated AML patients or primed in healthy donors. We describe the spontaneous appearance and persistence of NPM1-mutated-specific T cells, which may contribute to the maintenance of long-lasting remissions. Future studies are warranted to investigate the potential role of both autologous and allogeneic adoptive immunotherapy in NPM1-mutated AML patients.
Collapse
Affiliation(s)
- Fabio Forghieri
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Giovanni Riva
- Department of Laboratory Medicine and Pathology, Unità Sanitaria Locale, Modena, Italy
| | - Ivana Lagreca
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Patrizia Barozzi
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Daniela Vallerini
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Monica Morselli
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Ambra Paolini
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Paola Bresciani
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Elisabetta Colaci
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Monica Maccaferri
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Andrea Gilioli
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Vincenzo Nasillo
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Andrea Messerotti
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Valeria Pioli
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Laura Arletti
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Davide Giusti
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Francesca Bettelli
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Melania Celli
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Francesca Donatelli
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Giorgia Corradini
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Sabrina Basso
- Pediatric Hematology/Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy.,Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Antonella Gurrado
- Pediatric Hematology/Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy.,Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Monica Cellini
- Department of Medical and Surgical Sciences, Section of Pediatric Hemato-Oncology, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Unità Sanitaria Locale, Modena, Italy
| | - Roberto Marasca
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Franco Narni
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Maria Paola Martelli
- Institute of Hematology, Centro di Ricerca Emato-Oncologico, University of Perugia, Ospedale S. Maria della Misericordia, S. Andrea delle Fratte, Perugia, Italy
| | - Brunangelo Falini
- Institute of Hematology, Centro di Ricerca Emato-Oncologico, University of Perugia, Ospedale S. Maria della Misericordia, S. Andrea delle Fratte, Perugia, Italy
| | - Leonardo Potenza
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Patrizia Comoli
- Pediatric Hematology/Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy.,Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
125
|
The prognostic potential of monitoring disease dynamics in NPM1-positive acute myeloid leukemia. Leukemia 2019; 33:1531-1534. [PMID: 30675011 DOI: 10.1038/s41375-018-0371-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/07/2018] [Accepted: 12/13/2018] [Indexed: 11/08/2022]
|
126
|
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: 82] [Impact Index Per Article: 16.4] [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
|
127
|
Moors I, Vandepoele K, Philippé J, Deeren D, Selleslag D, Breems D, Straetmans N, Kerre T, Denys B. Clinical implications of measurable residual disease in AML: Review of current evidence. Crit Rev Oncol Hematol 2019; 133:142-148. [DOI: 10.1016/j.critrevonc.2018.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/30/2018] [Accepted: 11/23/2018] [Indexed: 02/08/2023] Open
|
128
|
Martignoles JA, Delhommeau F, Hirsch P. Genetic Hierarchy of Acute Myeloid Leukemia: From Clonal Hematopoiesis to Molecular Residual Disease. Int J Mol Sci 2018; 19:E3850. [PMID: 30513905 PMCID: PMC6321602 DOI: 10.3390/ijms19123850] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/25/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
Recent advances in the field of cancer genome analysis revolutionized the picture we have of acute myeloid leukemia (AML). Pan-genomic studies, using either single nucleotide polymorphism arrays or whole genome/exome next generation sequencing, uncovered alterations in dozens of new genes or pathways, intimately connected with the development of leukemia. From a simple two-hit model in the late nineties, we are now building clonal stories that involve multiple unexpected cellular functions, leading to full-blown AML. In this review, we will address several seminal concepts that result from these new findings. We will describe the genetic landscape of AML, the association and order of events that define multiple sub-entities, both in terms of pathogenesis and in terms of clinical practice. Finally, we will discuss the use of this knowledge in the settings of new strategies for the evaluation of measurable residual diseases (MRD), using clone-specific multiple molecular targets.
Collapse
Affiliation(s)
- Jean-Alain Martignoles
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Hématologie Biologique, F-75012 Paris, France.
| | - François Delhommeau
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Hématologie Biologique, F-75012 Paris, France.
| | - Pierre Hirsch
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Hématologie Biologique, F-75012 Paris, France.
| |
Collapse
|
129
|
Platzbecker U, Middeke JM, Sockel K, Herbst R, Wolf D, Baldus CD, Oelschlägel U, Mütherig A, Fransecky L, Noppeney R, Bug G, Götze KS, Krämer A, Bochtler T, Stelljes M, Groth C, Schubert A, Mende M, Stölzel F, Borkmann C, Kubasch AS, von Bonin M, Serve H, Hänel M, Dührsen U, Schetelig J, Röllig C, Kramer M, Ehninger G, Bornhäuser M, Thiede C. Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): an open-label, multicentre, phase 2 trial. Lancet Oncol 2018; 19:1668-1679. [DOI: 10.1016/s1470-2045(18)30580-1] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022]
|
130
|
Patkar N, Kodgule R, Kakirde C, Raval G, Bhanshe P, Joshi S, Chaudhary S, Badrinath Y, Ghoghale S, Kadechkar S, Khizer SH, Kannan S, Shetty D, Gokarn A, Punatkar S, Jain H, Bagal B, Menon H, Sengar M, Khattry N, Tembhare P, Subramanian P, Gujral S. Clinical impact of measurable residual disease monitoring by ultradeep next generation sequencing in NPM1 mutated acute myeloid leukemia. Oncotarget 2018; 9:36613-36624. [PMID: 30564301 PMCID: PMC6290958 DOI: 10.18632/oncotarget.26400] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/16/2018] [Indexed: 01/18/2023] Open
Abstract
Detection of measurable residual disease (MRD) by mutation specific techniques has prognostic relevance in NPM1 mutated AML (NPM1mut AML). However, the clinical utility of next generation sequencing (NGS) to detect MRD in AML remains unproven. We analysed the clinical significance of monitoring MRD using ultradeep NGS (NGS-MRD) and flow cytometry (FCM-MRD) in 137 samples obtained from 83 patients of NPM1mut AML at the end of induction (PI) and consolidation (PC). We could monitor 12 different types of NPM1 mutations at a sensitivity of 0.001% using NGS-MRD. We demonstrated a significant correlation between NGS-MRD and real time quantitative PCR (RQ-PCR). Based upon a one log reduction between PI and PC time points we could classify patients as NGS-MRD positive (<1log reduction) or negative (>1log reduction). NGS-MRD, FCM-MRD as well as DNMT3A mutations were predictive of inferior overall survival (OS) and relapse free survival (RFS). On a multivariate analysis NGS-MRD emerged as an independent, most important prognostic factor predictive of inferior OS (hazard ratio, 3.64; 95% confidence interval [CI] 1.58 to 8.37) and RFS (hazard ratio, 4.8; 95% CI:2.24 to 10.28). We establish that DNA based NPM1 NGS MRD is a highly useful test for prediction of relapse and survival in NPM1mut AML.
Collapse
Affiliation(s)
- Nikhil Patkar
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Rohan Kodgule
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India.,Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Chinmayee Kakirde
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Goutham Raval
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Prasanna Bhanshe
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Swapnali Joshi
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Shruti Chaudhary
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Y Badrinath
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Sitaram Ghoghale
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Shraddha Kadechkar
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Syed Hasan Khizer
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Sadhana Kannan
- Biostatistics, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Dhanalaxmi Shetty
- Dept of Cytogenetics, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Anant Gokarn
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Sachin Punatkar
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Hasmukh Jain
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Bhausaheb Bagal
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Hari Menon
- Haemato-Oncology, CyteCare Cancer Hospital, Bangalore, India
| | - Manju Sengar
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Navin Khattry
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Prashant Tembhare
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | | | - Sumeet Gujral
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| |
Collapse
|
131
|
[Expert consensus on the application of next-generation sequencing in hematological neoplasms (2018)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:881-886. [PMID: 30486580 PMCID: PMC7342343 DOI: 10.3760/cma.j.issn.0253-2727.2018.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Indexed: 01/15/2023]
|
132
|
Minimal/Measurable Residual Disease Monitoring in NPM1-Mutated Acute Myeloid Leukemia: A Clinical Viewpoint and Perspectives. Int J Mol Sci 2018; 19:ijms19113492. [PMID: 30404199 PMCID: PMC6274702 DOI: 10.3390/ijms19113492] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) with NPM1 gene mutations is currently recognized as a distinct entity, due to its unique biological and clinical features. We summarize here the results of published studies investigating the clinical application of minimal/measurable residual disease (MRD) in patients with NPM1-mutated AML, receiving either intensive chemotherapy or hematopoietic stem cell transplantation. Several clinical trials have so far demonstrated a significant independent prognostic impact of molecular MRD monitoring in NPM1-mutated AML and, accordingly, the Consensus Document from the European Leukemia Net MRD Working Party has recently recommended that NPM1-mutated AML patients have MRD assessment at informative clinical timepoints during treatment and follow-up. However, several controversies remain, mainly with regard to the most clinically significant timepoints and the MRD thresholds to be considered, but also with respect to the optimal source to be analyzed, namely bone marrow or peripheral blood samples, and the correlation of MRD with other known prognostic indicators. Moreover, we discuss potential advantages, as well as drawbacks, of newer molecular technologies such as digital droplet PCR and next-generation sequencing in comparison to conventional RQ-PCR to quantify NPM1-mutated MRD. In conclusion, further prospective clinical trials are warranted to standardize MRD monitoring strategies and to optimize MRD-guided therapeutic interventions in NPM1-mutated AML patients.
Collapse
|
133
|
Kayser S, Levis MJ. Clinical implications of molecular markers in acute myeloid leukemia. Eur J Haematol 2018; 102:20-35. [PMID: 30203623 DOI: 10.1111/ejh.13172] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
Abstract
The recently updated World Health Organization (WHO) Classification of myeloid neoplasms and leukemia reflects the fact that research in the underlying pathogenic mechanisms of acute myeloid leukemia (AML) has led to remarkable advances in our understanding of the disease. Gene mutations now allow us to explore the enormous diversity among cytogenetically defined subsets of AML, particularly the large subset of cytogenetically normal AML. Despite the progress in unraveling the tumor genome, only a small number of recurrent mutations have been incorporated into risk-stratification schemes and have been proven to be clinically relevant, targetable lesions. We here discuss the utility of molecular markers in AML in prognostication and treatment decision making, specifically highlighting the aberrations included in the current WHO classification.
Collapse
Affiliation(s)
- Sabine Kayser
- Department of Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark J Levis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
134
|
Schlenk RF, Jaramillo S, Müller-Tidow C. Improving consolidation therapy in acute myeloid leukemia - a tough nut to crack. Haematologica 2018; 103:1579-1581. [PMID: 30270203 DOI: 10.3324/haematol.2018.200485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Richard F Schlenk
- NCT-Trial Center, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany .,Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Germany
| | - Sonia Jaramillo
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Germany
| |
Collapse
|
135
|
Abstract
Interest in detecting minimal/measurable residual disease (MRD) in acute myeloid leukemia (AML) has been increasing, but numerous issues need to be addressed if MRD assessment is to be routinely incorporated into practice. Assays, their reliability, standardization, and availability all must be considered, and a strategy developed to eradicate residual leukemia. This paper reviews some issues surrounding the routine incorporation of MRD assessment into practice.
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, The University of Texas - M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX 77030, USA.
| |
Collapse
|
136
|
Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML. Blood 2018; 132:1703-1713. [PMID: 30190321 DOI: 10.1182/blood-2018-02-829911] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 08/24/2018] [Indexed: 12/14/2022] Open
Abstract
Molecular measurable residual disease (MRD) assessment is not established in approximately 60% of acute myeloid leukemia (AML) patients because of the lack of suitable markers for quantitative real-time polymerase chain reaction. To overcome this limitation, we established an error-corrected next-generation sequencing (NGS) MRD approach that can be applied to any somatic gene mutation. The clinical significance of this approach was evaluated in 116 AML patients undergoing allogeneic hematopoietic cell transplantation (alloHCT) in complete morphologic remission (CR). Targeted resequencing at the time of diagnosis identified a suitable mutation in 93% of the patients, covering 24 different genes. MRD was measured in CR samples from peripheral blood or bone marrow before alloHCT and identified 12 patients with persistence of an ancestral clone (variant allele frequency [VAF] >5%). The remaining 96 patients formed the final cohort of which 45% were MRD+ (median VAF, 0.33%; range, 0.016%-4.91%). In competing risk analysis, cumulative incidence of relapse (CIR) was higher in MRD+ than in MRD- patients (hazard ratio [HR], 5.58; P < .001; 5-year CIR, 66% vs 17%), whereas nonrelapse mortality was not significantly different (HR, 0.60; P = .47). In multivariate analysis, MRD positivity was an independent negative predictor of CIR (HR, 5.68; P < .001), in addition to FLT3-ITD and NPM1 mutation status at the time of diagnosis, and of overall survival (HR, 3.0; P = .004), in addition to conditioning regimen and TP53 and KRAS mutation status. In conclusion, NGS-based MRD is widely applicable to AML patients, is highly predictive of relapse and survival, and may help refine transplantation and posttransplantation management in AML patients.
Collapse
|
137
|
Schlenk RF, Jaramillo S, Müller-Tidow C. What's new in consolidation therapy in AML? Semin Hematol 2018; 56:96-101. [PMID: 30926097 DOI: 10.1053/j.seminhematol.2018.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/17/2022]
Abstract
Intensive induction chemotherapy followed by postremission treatment with either high-dose cytarabine-based regimens, autologous or allogeneic hematopoietic stem cell transplantation is still recognized as the main road toward cure in acute myeloid leukemia (AML). Pretreatment risk classification remains a key determinant of type and intensity of post-remission therapy. Still, high-dose cytarabine-based consolidation therapy is a cornerstone of postremission therapy with some recent adjustments regarding dosage and schedule. Current approvals of midostaurin, gemtuzumab ozogamicin, CPX-351, and ivosidenib as well as enasidenib comprise induction as well as consolidation therapy. In recent years measurable residual disease assessment is increasingly used to dynamically fine tune treatment during postremission treatment.
Collapse
Affiliation(s)
- Richard F Schlenk
- NCT-Trial Center, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany; Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany.
| | - Sonia Jaramillo
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
138
|
Methods and role of minimal residual disease after stem cell transplantation. Bone Marrow Transplant 2018; 54:681-690. [PMID: 30116018 DOI: 10.1038/s41409-018-0307-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/28/2018] [Accepted: 06/13/2018] [Indexed: 11/08/2022]
Abstract
Relapse is the major cause of treatment failure after stem cell transplantation. Despite the fact that relapses occurred even if transplantation was performed in complete remission, it is obvious that minimal residual disease is present though not morphologically evident. Since adaptive immunotherapy by donor lymphocyte infusion or other novel cell therapies as well as less toxic drugs, which can be used after transplantation, the detection of minimal residual disease (MRD) has become a clinical important variable for outcome. Besides the increasing options to treat MRD, the most advanced technologies currently allow to detect residual malignant cells with a sensitivity of 10-5 to 10-6.Under the patronage of the European Society for Blood and Marrow Transplantation (EBMT) and the American Society for Blood and Marrow Transplantation (ASBMT) the 3rd workshop was held on 4/5 November 2016 in Hamburg/Germany, with the aim to present an up-to-date status of epidemiology and biology of relapse and to summarize the currently available options to prevent and treat post-transplant relapse. Here the current methods and role of minimal residual disease for myeloid and lymphoid malignancies are summarized.
Collapse
|
139
|
Onecha E, Linares M, Rapado I, Ruiz-Heredia Y, Martinez-Sanchez P, Cedena T, Pratcorona M, Oteyza JP, Herrera P, Barragan E, Montesinos P, Vela JAG, Magro E, Anguita E, Figuera A, Riaza R, Martinez-Barranco P, Sanchez-Vega B, Nomdedeu J, Gallardo M, Martinez-Lopez J, Ayala R. A novel deep targeted sequencing method for minimal residual disease monitoring in acute myeloid leukemia. Haematologica 2018; 104:288-296. [PMID: 30093399 PMCID: PMC6355493 DOI: 10.3324/haematol.2018.194712] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
A high proportion of patients with acute myeloid leukemia who achieve minimal residual disease negative status ultimately relapse because a fraction of pathological clones remains undetected by standard methods. We designed and validated a high-throughput sequencing method for minimal residual disease assessment of cell clonotypes with mutations of NPM1, IDH1/2 and/or FLT3-single nucleotide variants. For clinical validation, 106 follow-up samples from 63 patients in complete remission were studied by sequencing, evaluating the level of mutations detected at diagnosis. The predictive value of minimal residual disease status by sequencing, multiparameter flow cytometry, or quantitative polymerase chain reaction analysis was determined by survival analysis. The sequencing method achieved a sensitivity of 10−4 for single nucleotide variants and 10−5 for insertions/deletions and could be used in acute myeloid leukemia patients who carry any mutation (86% in our diagnostic data set). Sequencing–determined minimal residual disease positive status was associated with lower disease-free survival (hazard ratio 3.4, P=0.005) and lower overall survival (hazard ratio 4.2, P<0.001). Multivariate analysis showed that minimal residual disease positive status determined by sequencing was an independent factor associated with risk of death (hazard ratio 4.54, P=0.005) and the only independent factor conferring risk of relapse (hazard ratio 3.76, P=0.012). This sequencing-based method simplifies and standardizes minimal residual disease evaluation, with high applicability in acute myeloid leukemia. It is also an improvement upon flow cytometry- and quantitative polymerase chain reaction-based prediction of outcomes of patients with acute myeloid leukemia and could be incorporated in clinical settings and clinical trials.
Collapse
Affiliation(s)
- Esther Onecha
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid
| | - Maria Linares
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid
| | - Inmaculada Rapado
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid
| | - Yanira Ruiz-Heredia
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid
| | | | - Teresa Cedena
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid.,Complutense University, Madrid
| | - Marta Pratcorona
- Hematology Department, Hospital Santa Creu i Sant Pau, Barcelona
| | | | - Pilar Herrera
- Hematology Department, Hospital Universitario Ramon y Cajal, Madrid
| | - Eva Barragan
- Complutense University, Madrid.,Hematology Department, Hospital Universitario La Fe, Valencia
| | - Pau Montesinos
- Complutense University, Madrid.,Hematology Department, Hospital Universitario La Fe, Valencia
| | | | - Elena Magro
- Hematology Department, Hospital Universitario Principe de Asturias, Madrid
| | - Eduardo Anguita
- Hematology Department, Hospital Clínico San Carlos, IdISSC, UCM, Madrid
| | - Angela Figuera
- Hematology Department, Hospital Universitario de la Princesa, Madrid
| | - Rosalia Riaza
- Hematology Department, Hospital Universitario Severo Ochoa, Madrid
| | | | - Beatriz Sanchez-Vega
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid
| | - Josep Nomdedeu
- Hematology Department, Hospital Santa Creu i Sant Pau, Barcelona
| | - Miguel Gallardo
- Hematological Malignancies Clinical Research Unit, CNIO, Madrid
| | - Joaquin Martinez-Lopez
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid.,Complutense University, Madrid
| | - Rosa Ayala
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid .,Hematological Malignancies Clinical Research Unit, CNIO, Madrid.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid.,Complutense University, Madrid
| |
Collapse
|
140
|
Molecular remission at the end of treatment is a necessary goal for a good outcome in ELN favorable-risk acute myeloid leukemia: a real-life analysis on 201 patients by the Rete Ematologica Lombarda network. Ann Hematol 2018; 97:2107-2115. [PMID: 30009341 DOI: 10.1007/s00277-018-3424-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/27/2018] [Indexed: 10/28/2022]
Abstract
Favorable acute myeloid leukemia (AML) patients (pts.) demonstrate a relatively good outcome with standard induction; thus, pts. are generally not addressed to allogeneic transplant in first remission. However, it is not clear if also in a real-life setting, the outcome is homogeneous in the different favorable molecular groups and which are the parameters significantly associated to an increased relapse risk, useful to suggest the need of an intensified approach. In order to clarify this point, we collected clinical data on consecutive unselected AML pts. assigned to favorable category (modified ELN 2010 due to the inclusion of double-mutated CEBPA-positive cases), diagnosed and treated in six centers of the Italian network Rete Ematologica Lombarda (REL) from 2007 to 2015. We assessed response (CR, mCR), relapse rate (CIR), and outcome (OS, DFS) after first-line treatment. A total of 201 pts. was studied and the analysis was performed globally and in each molecular group: t(8;21)(q22;q22)/RUNX1-RUNX1T1 (30 pts., 14.9%), inv. (16)(p13q22) or t(16;16)(p13q22)/CBFB-MIH11 (35 pts., 17.4%), normal karyotype and mutated NPM1 and negative FLT3-ITD (116 pts., 57.7%) or double-mutated CEBPA (CEBPAdm) (20 pts., 10%). Complete remission (CR) was obtained in 188 pts. (93.5%), molecular CR (mCR) in 114 (67.5%); After a median follow-up of 2.4 years, cumulative incidence of relapse (CIR) was documented in 78 of 188 responding pts. (41%) after a median time of 11.3 months. CIR was higher in the CBFB-MIH11 group, in pts. achieving only a hematological response without mCR (72.1 vs 28.1%, p < 0.001), in older pts. and it resulted independently associated with a lower median cytarabine cumulative dose (CCD). Median OS was not reached: after 5 years it was 66.3%, and median DFS was 5.3 years, both without difference among groups. Molecular CR reached at any time, during or after the end of first-line treatment, was significantly associated with better DFS, and in particular, mCR assessed at the end of treatment was confirmed in multivariate analysis as an independent prognostic factor both for DFS and OS. In conclusion, the present study confirms in a real-life context the overall good prognosis of favorable-risk AML; the achievement of any molecular negativity during first-line treatment, particularly when assessed at the end of treatment, is associated with lower relapse and better survival. Increasing age at diagnosis has a negative prognostic impact, while CCD higher than 18 g/sqm is associated with better outcome.
Collapse
|
141
|
Abstract
The presence of measurable ("minimal") residual disease (MRD) after induction and/or consolidation chemotherapy is a significant risk factor for relapse in patients with acute myeloid leukemia (AML). In recognition of the clinical significance of AML MRD, the European LeukemiaNet (ELN) recently recommended the establishment of CR-MRDNegative as a separate category of treatment response. This recommendation represents a major milestone in the integration of AML MRD testing in standard clinical practice. This review article summarizes the methodologies employed in AML MRD detection and their application in clinical studies that provide evidence supporting the clinical utility of AML MRD testing. Future MRD evaluations in AML likely will require an integrated approach combining multi-parameter flow cytometry and high-sensitivity molecular techniques applied to time points during and after completion of therapy in order to provide the most accurate and comprehensive assessment of treatment response.
Collapse
|
142
|
Abstract
PURPOSE OF REVIEW Assessment of measurable residual disease (MRD) after treatment can identify patients with acute myeloid leukemia (AML) that are at high risk of poor outcomes. However, there is no consensus yet regarding a standardized approach to measuring MRD that is most clinically meaningful. We review multiparameter flow cytometry (MFC) and reverse transcriptase polymerase chain reaction (RT-PCR), and discuss a framework for assessing remission MRD using next-generation sequencing (NGS). RECENT FINDINGS MFC and RT-PCR may not fully capitalize on the major advances that have been made in characterizing the genetic landscape of AML, which has offered insight into the biological and clinical implications of clonal genetic architecture. NGS has increasingly been shown to provide a qualitative and quantitative assessment of MRD with significant prognostic implications. The assessment of clonal architecture by NGS may complement or extend existing approaches for MRD monitoring. Long-term serial monitoring of diagnostic, remission, and relapse samples with clinical correlation will need to be performed in order to determine the impact of various MRD patterns using this technique.
Collapse
|
143
|
Bacher U, Porret N, Joncourt R, Sanz J, Aliu N, Wiedemann G, Jeker B, Banz Y, Pabst T. Pitfalls in the molecular follow up of NPM1 mutant acute myeloid leukemia. Haematologica 2018; 103:e486-e488. [PMID: 29903758 DOI: 10.3324/haematol.2018.192104] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Ulrike Bacher
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Switzerland.,Center of Laboratory Medicine (ZLM) Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Naomi Porret
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Raphael Joncourt
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Javier Sanz
- Division of Human Genetics, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Nijas Aliu
- Division of Human Genetics, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Gertrud Wiedemann
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Barbara Jeker
- Department of Medical Oncology, Inselspital, Bern University Hospital, Switzerland
| | - Yara Banz
- Institute of Pathology, University of Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, Bern University Hospital, Switzerland
| |
Collapse
|
144
|
Bill M, Grimm J, Jentzsch M, Kloss L, Goldmann K, Schulz J, Beinicke S, Häntschel J, Cross M, Vucinic V, Pönisch W, Behre G, Franke GN, Lange T, Niederwieser D, Schwind S. Digital droplet PCR-based absolute quantification of pre-transplant NPM1 mutation burden predicts relapse in acute myeloid leukemia patients. Ann Hematol 2018; 97:1757-1765. [DOI: 10.1007/s00277-018-3373-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 05/12/2018] [Indexed: 12/24/2022]
|
145
|
Coustan-Smith E, Song G, Shurtleff S, Yeoh AEJ, Chng WJ, Chen SP, Rubnitz JE, Pui CH, Downing JR, Campana D. Universal monitoring of minimal residual disease in acute myeloid leukemia. JCI Insight 2018; 3:98561. [PMID: 29720577 DOI: 10.1172/jci.insight.98561] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/28/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Optimal management of acute myeloid leukemia (AML) requires monitoring of treatment response, but minimal residual disease (MRD) may escape detection. We sought to identify distinctive features of AML cells for universal MRD monitoring. METHODS We compared genome-wide gene expression of AML cells from 157 patients with that of normal myeloblasts. Markers encoded by aberrantly expressed genes, including some previously associated with leukemia stem cells, were studied by flow cytometry in 240 patients with AML and in nonleukemic myeloblasts from 63 bone marrow samples. RESULTS Twenty-two (CD9, CD18, CD25, CD32, CD44, CD47, CD52, CD54, CD59, CD64, CD68, CD86, CD93, CD96, CD97, CD99, CD123, CD200, CD300a/c, CD366, CD371, and CX3CR1) markers were aberrantly expressed in AML. Leukemia-associated profiles defined by these markers extended to immature CD34+CD38- AML cells; expression remained stable during treatment. The markers yielded MRD measurements matching those of standard methods in 208 samples from 52 patients undergoing chemotherapy and revealed otherwise undetectable MRD. They allowed MRD monitoring in 129 consecutive patients, yielding prognostically significant results. Using a machine-learning algorithm to reduce high-dimensional data sets to 2-dimensional data, the markers allowed a clear visualization of MRD and could detect 1 leukemic cell among more than 100,000 normal cells. CONCLUSION The markers uncovered in this study allow universal and sensitive monitoring of MRD in AML. In combination with contemporary analytical tools, the markers improve the discrimination between leukemic and normal cells, thus facilitating data interpretation and, hence, the reliability of MRD results. FUNDING National Cancer Institute (CA60419 and CA21765); American Lebanese Syrian Associated Charities; National Medical Research Council of Singapore (1299/2011); Viva Foundation for Children with Cancer, Children's Cancer Foundation, Tote Board & Turf Club, and Lee Foundation of Singapore.
Collapse
Affiliation(s)
| | - Guangchun Song
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sheila Shurtleff
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Allen Eng-Juh Yeoh
- Department of Pediatrics, National University of Singapore, Singapore.,National University Cancer Institute, Singapore, National University of Singapore, Singapore
| | - Wee Joo Chng
- National University Cancer Institute, Singapore, National University of Singapore, Singapore
| | - Siew Peng Chen
- Department of Pediatrics, National University of Singapore, Singapore
| | - Jeffrey E Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.,University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ching-Hon Pui
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.,University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James R Downing
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.,University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dario Campana
- Department of Pediatrics, National University of Singapore, Singapore.,National University Cancer Institute, Singapore, National University of Singapore, Singapore
| |
Collapse
|
146
|
Coltoff A, Houldsworth J, Keyzner A, Renteria AS, Mascarenhas J. Role of minimal residual disease in the management of acute myeloid leukemia-a case-based discussion. Ann Hematol 2018; 97:1155-1167. [PMID: 29704019 DOI: 10.1007/s00277-018-3330-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/04/2018] [Indexed: 01/04/2023]
Abstract
AML is stratified into risk-categories based on cytogenetic and molecular features that prognosticate survival and facilitate treatment algorithms, though there is still significant heterogeneity within risk groupings with regard to risk of relapse and prognosis. The ambiguity regarding prognosis is due in large part to the relatively outdated criteria used to determine response to therapy. Whereas risk assessment has evolved to adopt cytogenetic and molecular profiling, response criteria are still largely determined by bone marrow morphologic assessment and peripheral cell count recovery. Minimal residual disease refers to the detection of a persistent population of leukemic cells below the threshold for morphologic CR determination. MRD assessment represents standard of care for ALL and PML, but concerns over prognostic capability and standardization have limited its use in AML. However, recent advancements in MRD assessment and research supporting the use of MRD assessment in AML require the reconsideration and review of this clinical tool in this disease entity. This review article will first compare and contrast the major modalities used to assess MRD in AML, such as RQ-PCR and flow cytometry, as well as touching upon newer technologies such as next-generation sequencing and digital droplet PCR. The majority of the article will discuss the evidence supporting the use of MRD assessment to prognosticate disease at various time points during treatment, and review the limited number of studies that have incorporated MRD assessment into novel treatment algorithms for AML. The article concludes by discussing the current major limitations to the implementation of MRD assessment in this disease. The manuscript is bookended by a clinical vignette that highlights the need for further research and refinement of this clinical tool.
Collapse
Affiliation(s)
- A Coltoff
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Houldsworth
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A Keyzner
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A S Renteria
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Adult Leukemia Program, Myeloproliferative Disorders Clinical Research Program, Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY, 10029, USA.
| |
Collapse
|
147
|
Morita K, Kantarjian HM, Wang F, Yan Y, Bueso-Ramos C, Sasaki K, Issa GC, Wang S, Jorgensen J, Song X, Zhang J, Tippen S, Thornton R, Coyle M, Little L, Gumbs C, Pemmaraju N, Daver N, DiNardo CD, Konopleva M, Andreeff M, Ravandi F, Cortes JE, Kadia T, Jabbour E, Garcia-Manero G, Patel KP, Futreal PA, Takahashi K. Clearance of Somatic Mutations at Remission and the Risk of Relapse in Acute Myeloid Leukemia. J Clin Oncol 2018; 36:1788-1797. [PMID: 29702001 DOI: 10.1200/jco.2017.77.6757] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose The aim of the current study was to determine whether the degree of mutation clearance at remission predicts the risk of relapse in patients with acute myeloid leukemia (AML). Patients and Methods One hundred thirty-one previously untreated patients with AML who received intensive induction chemotherapy and attained morphologic complete remission (CR) at day 30 were studied. Pretreatment and CR bone marrow were analyzed using targeted capture DNA sequencing. We analyzed the association between mutation clearance (MC) on the basis of variant allele frequency (VAF) at CR (MC2.5: if the VAF of residual mutations was < 2.5%; MC1.0: if the VAF was < 1%; and complete MC [CMC]: if no detectable residual mutations) and event-free survival, overall survival (OS), and cumulative incidence of relapse (CIR). Results MC1.0 and CMC were associated with significantly better OS (2-year OS: 75% v 61% in MC1.0 v non-MC1.0; P = .0465; 2-year OS: 77% v 60% in CMC v non-CMC; P = .0303) and lower CIR (2-year CIR: 26% v 46% in MC1.0 v non-MC 1.0; P = .0349; 2 year-CIR: 24% v 46% in CMC v non-CMC; P = .03), whereas there was no significant difference in any of the above outcomes by MC2.5. Multivariable analysis adjusting for age, cytogenetic risk, allogeneic stem-cell transplantation, and flow cytometry-based minimal residual disease revealed that patients with CMC had significantly better event-free survival (hazard ratio [HR], 0.43; P = .0083), OS (HR, 0.47; P = .04), and CIR (HR, 0.27; P < .001) than did patients without CMC. These prognostic associations were stronger when preleukemic mutations, such as DNMT3A, TET2, and ASXL1, were removed from the analysis. Conclusion Clearance of somatic mutation at CR, particularly in nonpreleukemic genes, was associated with significantly better survival and less risk of relapse. Somatic mutations in nonpreleukemic genes may function as a molecular minimal residual disease marker in AML.
Collapse
Affiliation(s)
- Kiyomi Morita
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Hagop M Kantarjian
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Feng Wang
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Yuanqing Yan
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Carlos Bueso-Ramos
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Koji Sasaki
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Ghayas C Issa
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Sa Wang
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Jeffrey Jorgensen
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Xingzhi Song
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Jianhua Zhang
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Samantha Tippen
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Rebecca Thornton
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Marcus Coyle
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Latasha Little
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Curtis Gumbs
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Naveen Pemmaraju
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Naval Daver
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Courtney D DiNardo
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Marina Konopleva
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Michael Andreeff
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Farhad Ravandi
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Jorge E Cortes
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Tapan Kadia
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Elias Jabbour
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Guillermo Garcia-Manero
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Keyur P Patel
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - P Andrew Futreal
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| | - Koichi Takahashi
- Kiyomi Morita, Hagop M. Kantarjian, Feng Wang, Yuanqing Yan, Carlos Bueso-Ramos, Koji Sasaki, Ghayas C. Issa, Sa Wang, Jeffrey Jorgensen, Xingzhi Song, Jianhua Zhang, Samantha Tippen, Rebecca Thornton, Marcus Coyle, Latasha Little, Curtis Gumbs, Naveen Pemmaraju, Naval Daver, Courtney D. DiNardo, Marina Konopleva, Michael Andreeff, Farhad Ravandi, Jorge E. Cortes, Tapan Kadia, Elias Jabbour, Guillermo Garcia-Manero, Keyur P. Patel, P. Andrew Futreal, and Koichi Takahash, The University of Texas MD Anderson Cancer Center, Houston, TX; Kiyomi Morita, The University of Tokyo, Tokyo; and Koichi Takahashi, Kyoto University, Kyoto, Japan
| |
Collapse
|
148
|
Dutta D, Chong NS, Lim SH. Endogenous volatile organic compounds in acute myeloid leukemia: origins and potential clinical applications. J Breath Res 2018; 12:034002. [PMID: 29463782 DOI: 10.1088/1752-7163/aab108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Not unlike many cancer types, acute myeloid leukemia (AML) exhibits many metabolic changes and reprogramming, causing changes in lipid metabolism. Some of the distinct molecular abnormalities associated with AML also modify the metabolic changes. Both processes result in changes in the production of endogenous volatile organic compounds (VOCs). The increasing availability of highly sensitive methods for detecting trace chemicals provides the opportunity to investigate the role of patient-specific VOC finger-prints as biomarkers for detecting early relapse or minimal residual disease in AML. Since VOC production is reliant on metabolic activities, when combined with currently available methods, VOC analysis may identify within a group of patients with flow cytometric or molecular evidence of residual disease those most at risk for disease relapse.
Collapse
Affiliation(s)
- Dibyendu Dutta
- Department of Professional Sciences, Middle Tennessee State University, Murfreesboro, Tennessee, United States of America
| | | | | |
Collapse
|
149
|
Chen J, Yang L, Fan Y, Xu Y, Han Y, Tang X, Qiu H, Fu C, Miao M, Chen F, Wu D. Comparison of Autologous Stem Cell Transplantation versus Haploidentical Donor Stem Cell Transplantation for Favorable- and Intermediate-Risk Acute Myeloid Leukemia Patients in First Complete Remission. Biol Blood Marrow Transplant 2018; 24:779-788. [DOI: 10.1016/j.bbmt.2017.12.796] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/18/2017] [Indexed: 01/03/2023]
|
150
|
Jongen-Lavrencic M, Grob T, Hanekamp D, Kavelaars FG, Al Hinai A, Zeilemaker A, Erpelinck-Verschueren CAJ, Gradowska PL, Meijer R, Cloos J, Biemond BJ, Graux C, van Marwijk Kooy M, Manz MG, Pabst T, Passweg JR, Havelange V, Ossenkoppele GJ, Sanders MA, Schuurhuis GJ, Löwenberg B, Valk PJM. Molecular Minimal Residual Disease in Acute Myeloid Leukemia. N Engl J Med 2018; 378:1189-1199. [PMID: 29601269 DOI: 10.1056/nejmoa1716863] [Citation(s) in RCA: 539] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Patients with acute myeloid leukemia (AML) often reach complete remission, but relapse rates remain high. Next-generation sequencing enables the detection of molecular minimal residual disease in virtually every patient, but its clinical value for the prediction of relapse has yet to be established. METHODS We conducted a study involving patients 18 to 65 years of age who had newly diagnosed AML. Targeted next-generation sequencing was carried out at diagnosis and after induction therapy (during complete remission). End points were 4-year rates of relapse, relapse-free survival, and overall survival. RESULTS At least one mutation was detected in 430 out of 482 patients (89.2%). Mutations persisted in 51.4% of those patients during complete remission and were present at various allele frequencies (range, 0.02 to 47%). The detection of persistent DTA mutations (i.e., mutations in DNMT3A, TET2, and ASXL1), which are often present in persons with age-related clonal hematopoiesis, was not correlated with an increased relapse rate. After the exclusion of persistent DTA mutations, the detection of molecular minimal residual disease was associated with a significantly higher relapse rate than no detection (55.4% vs. 31.9%; hazard ratio, 2.14; P<0.001), as well as with lower rates of relapse-free survival (36.6% vs. 58.1%; hazard ratio for relapse or death, 1.92; P<0.001) and overall survival (41.9% vs. 66.1%; hazard ratio for death, 2.06; P<0.001). Multivariate analysis confirmed that the persistence of non-DTA mutations during complete remission conferred significant independent prognostic value with respect to the rates of relapse (hazard ratio, 1.89; P<0.001), relapse-free survival (hazard ratio for relapse or death, 1.64; P=0.001), and overall survival (hazard ratio for death, 1.64; P=0.003). A comparison of sequencing with flow cytometry for the detection of residual disease showed that sequencing had significant additive prognostic value. CONCLUSIONS Among patients with AML, the detection of molecular minimal residual disease during complete remission had significant independent prognostic value with respect to relapse and survival rates, but the detection of persistent mutations that are associated with clonal hematopoiesis did not have such prognostic value within a 4-year time frame. (Funded by the Queen Wilhelmina Fund Foundation of the Dutch Cancer Society and others.).
Collapse
Affiliation(s)
- Mojca Jongen-Lavrencic
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Tim Grob
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Diana Hanekamp
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - François G Kavelaars
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Adil Al Hinai
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Annelieke Zeilemaker
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Claudia A J Erpelinck-Verschueren
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Patrycja L Gradowska
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Rosa Meijer
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Jacqueline Cloos
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Bart J Biemond
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Carlos Graux
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Marinus van Marwijk Kooy
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Markus G Manz
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Thomas Pabst
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Jakob R Passweg
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Violaine Havelange
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Gert J Ossenkoppele
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Mathijs A Sanders
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Gerrit J Schuurhuis
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Bob Löwenberg
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| | - Peter J M Valk
- From the Department of Hematology (M.J.-L., T.G., F.G.K., A.H., A.Z., C.A.J.E.-V., M.A.S., B.L., P.J.M.V.) and HOVON Data Center, Department of Hematology (P.L.G., R.M.), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Department of Hematology, VU University Medical Center (D.H., J.C., G.J.O., G.J.S.), and the Department of Hematology, Academic Medical Center (B.J.B.), Amsterdam, and Isala Hospital, Zwolle (M.M.K.) - all in the Netherlands; UCL Namur (Godinne), Yvoir (C.G.), and the Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels (V.H.) - both in Belgium; and the Department of Hematology, University Hospital Zurich, Zurich (M.G.M.), University Hospital, Bern (T.P.), and the Division of Hematology, University Hospital Basel, Basel (J.R.P.) - all in Switzerland
| |
Collapse
|