151
|
Boyiadzis MM, Aksentijevich I, Arber DA, Barrett J, Brentjens RJ, Brufsky J, Cortes J, De Lima M, Forman SJ, Fuchs EJ, Fukas LJ, Gore SD, Litzow MR, Miller JS, Pagel JM, Waller EK, Tallman MS. The Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of acute leukemia. J Immunother Cancer 2020; 8:jitc-2020-000810. [PMID: 33077513 PMCID: PMC7574947 DOI: 10.1136/jitc-2020-000810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2020] [Indexed: 12/29/2022] Open
Abstract
Acute leukemia is a constellation of rapidly progressing diseases that affect a wide range of patients regardless of age or gender. Traditional treatment options for patients with acute leukemia include chemotherapy and hematopoietic cell transplantation. The advent of cancer immunotherapy has had a significant impact on acute leukemia treatment. Novel immunotherapeutic agents including antibody-drug conjugates, bispecific T cell engagers, and chimeric antigen receptor T cell therapies have efficacy and have recently been approved by the US Food and Drug Administration (FDA) for the treatment of patients with acute leukemia. The Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop a clinical practice guideline composed of consensus recommendations on immunotherapy for the treatment of acute lymphoblastic leukemia and acute myeloid leukemia.
Collapse
Affiliation(s)
- Michael M Boyiadzis
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - John Barrett
- Stem Cell Allotransplantation Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jill Brufsky
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jorge Cortes
- Department of Medicine, Division of Hematology/Oncology, Georgia Cancer Center, Augusta, Georgia, USA
| | - Marcos De Lima
- Division of Hematology, University Hospitals of Cleveland and Case Western Reserve University, Cleveland, Ohio, USA
| | - Stephen J Forman
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Ephraim J Fuchs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linda J Fukas
- Clinical Research Services, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Steven D Gore
- Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark R Litzow
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey S Miller
- Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - John M Pagel
- Center for Blood Disorders and Stem Cell Transplantation, Swedish Cancer Institute, Seattle, Washington, USA
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Martin S Tallman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
152
|
Valkova V, Vydra J, Markova M, Cerovska E, Vrana M, Marinov I, Cechova H, Cetkovsky P, Vitek A, Salek C. WT1 Gene Expression in Peripheral Blood Before and After Allogeneic Stem Cell Transplantation is a Clinically Relevant Prognostic Marker in AML - A Single-center 14-year Experience. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:e145-e151. [PMID: 33160932 DOI: 10.1016/j.clml.2020.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND This work summarizes our experience with WT1 monitoring before and after allogeneic hematopoietic stem cell transplantation (allo-HSCT). PATIENTS AND METHODS The expression of WT1 gene was measured by real-time polymerase chain reaction in peripheral blood according to the European Leukemia Net recommendations. Between May 2005 and August 2019, we analyzed 147 consecutive patients with acute myeloid leukemia with high WT1 expression at diagnosis, transplanted in first (CR1) or second (CR2) complete remission. RESULTS At the time of allo-HSCT, 107 patients had WT1-normal expression (WT1 ≤ 50 copies), and 40 patients had WT1-high expression. The median follow-up was 21 months. The estimated 5-year overall survival and event-free survival was significantly better in the WT1-normal cohort (65% and 57% vs. 37% and 25%; P = .0003 and P < .0001, respectively) and 5-year cumulative incidence of relapse was significantly lower in the WT1-normal group (19% vs. 53%; P < .0001). Five-year non-relapse mortality was not significantly different (20% and 23%). Multivariate analysis revealed WT1-high expression and acute graft-versus-host disease grade 3/4 as significantly negative prognostic factors for OS. Overall, 49 patients developed WT1 molecular relapse in the post-transplant period; in 14 cases, the therapeutic intervention was done. In all but 1 relapsed patient where WT1 minimal residual disease (MRD) was monitored (38 patients), we detected WT1-high levels (sensitivity of 97%). CONCLUSION The results of the analysis confirmed our previous experience that WT1 status before allo-HSCT is a strong prognostic factor for both OS and relapse risk. In addition, we confirmed the usefulness of this marker for MRD monitoring after allo-HSCT. The main advantage is the possibility of frequent MRD monitoring in peripheral blood and early bone marrow examination based on WT1-high expression.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/metabolism
- Disease-Free Survival
- Feasibility Studies
- Female
- Follow-Up Studies
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Graft vs Host Disease/diagnosis
- Graft vs Host Disease/epidemiology
- Hematopoietic Stem Cell Transplantation/adverse effects
- Humans
- Incidence
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/genetics
- Neoplasm, Residual
- Prognosis
- Risk Assessment/methods
- Risk Factors
- Severity of Illness Index
- WT1 Proteins/blood
- WT1 Proteins/metabolism
- Young Adult
Collapse
Affiliation(s)
- Veronika Valkova
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Jan Vydra
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Markova
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ela Cerovska
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Milena Vrana
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Iuri Marinov
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hana Cechova
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petr Cetkovsky
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Antonin Vitek
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cyril Salek
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
153
|
Guolo F, Fianchi L, Minetto P, Clavio M, Gottardi M, Galimberti S, Rizzuto G, Rondoni M, Bertani G, Dargenio M, Bilio A, Scappini B, Zappasodi P, Scattolin AM, Grimaldi F, Pietrantuono G, Musto P, Cerrano M, D'Ardia S, Audisio E, Cignetti A, Pasciolla C, Pavesi F, Candoni A, Gurreri C, Morselli M, Alati C, Fracchiolla N, Rossi G, Caizzi M, Carnevale-Schianca F, Tafuri A, Rossi G, Ferrara F, Pagano L, Lemoli RM. CPX-351 treatment in secondary acute myeloblastic leukemia is effective and improves the feasibility of allogeneic stem cell transplantation: results of the Italian compassionate use program. Blood Cancer J 2020; 10:96. [PMID: 33024084 PMCID: PMC7538937 DOI: 10.1038/s41408-020-00361-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/19/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
Secondary acute myeloid leukemia (sAML) poorly responds to conventional treatments and allogeneic stem cell transplantation (HSCT). We evaluated toxicity and efficacy of CPX-351 in 71 elderly patients (median age 66 years) with sAML enrolled in the Italian Named (Compassionate) Use Program. Sixty days treatment-related mortality was 7% (5/71). The response rate at the end of treatment was: CR/CRi in 50/71 patients (70.4%), PR in 6/71 (8.5%), and NR in 10/71 (19.7%). After a median follow-up of 11 months relapse was observed in 10/50 patients (20%) and 12 months cumulative incidence of relapse (CIR) was 23.6%. Median duration of response was not reached. In competing risk analysis, CIR was reduced when HSCT was performed in first CR (12 months CIR of 5% and 37.4%, respectively, for patients receiving (=20) or not (=30) HSCT, p = 0.012). Twelve-months OS was 68.6% (median not reached). In landmark analysis, HSCT in CR1 was the only significant predictor of longer survival (12 months OS of 100 and 70.5%, for patients undergoing or not HSCT in CR1, respectively, p = 0.011). In conclusion, we extend to a real-life setting, the notion that CPX is an effective regimen for high risk AML patients and may improve the results of HSCT.
Collapse
Affiliation(s)
- Fabio Guolo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy. .,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy.
| | - Luana Fianchi
- Istituto di Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Paola Minetto
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Marino Clavio
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Sara Galimberti
- UO Ematologia, Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Giuliana Rizzuto
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Michela Rondoni
- U.O.C. di Ematologia, Azienda Unità Sanitaria Locale della Romagna, Ravenna, Italy
| | - Giambattista Bertani
- S.C. Ematologia, ASST Grande Ospedale Metropolitano, Niguarda Ca' Granda Milano, Milan, Italy
| | | | | | - Barbara Scappini
- Dipartimento di Oncologia-SODc Ematologia, Azienda Ospedaliero - Universitaria Careggi, Florence, Italy
| | - Patrizia Zappasodi
- Clinica Ematologica, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Francesco Grimaldi
- Dipartimento di Medicina Clinica e Chirurgia, AOU Federico II di Napoli, Naples, Italy
| | | | - Pellegrino Musto
- IRCCS Centro Oncologico della Basilicata, Rionero in Vulture, Potenza, Italy.,Unit of Hematology and Stem Cell Transplantation, AOU Policlinico Consorziale, "Aldo Moro" University, Bari, Italy
| | - Marco Cerrano
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Turin, Italy
| | - Stefano D'Ardia
- Institute for Cancer Research and Treatment, University of Turin-School of Medicine, Turin, Italy
| | - Ernesta Audisio
- S.C. Ematologia2, Dipartimento di Ematologia e Oncologia, AO Città della Salute e della Scienza di Torino, Turin, Italy
| | - Alessandro Cignetti
- Divisione Universitaria di Ematologia e Terapie Cellulari, A.O. Ordine Mauriziano, Turin, Italy
| | | | - Francesca Pavesi
- Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Candoni
- Clinica Ematologica, Centro Trapianti e Terapie Cellulari, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Carmela Gurreri
- U.O. Ematologia ed Immunologia Clinica, Azienda Ospedaliera di Padova, Padova, Italy
| | - Monica Morselli
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | | | - Nicola Fracchiolla
- Oncoematologia, IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan, Italy
| | - Giovanni Rossi
- U.O. Ematologia, Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo, Foggia, Italy
| | - Manuela Caizzi
- S.C. Ematologia Azienda Sanitaria Universitaria Integrata di Trieste, Ospedale Maggiore, Trieste, Italy
| | - Fabrizio Carnevale-Schianca
- Medical Oncology, Hematopoietic Stem Cells Unit, Turin Metropolitan Transplant Center, Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
| | - Agostino Tafuri
- Department of Clinical and Molecular Medicine & Hematology, Sant'Andrea - University Hospital - Sapienza - University of Rome, Rome, Italy
| | - Giuseppe Rossi
- SC Ematologia e Dipartimento di Oncologia Clinica, A.O. Spedali Civili, Brescia, Italy
| | | | - Livio Pagano
- Istituto di Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Roberto Massimo Lemoli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| |
Collapse
|
154
|
Nagler A, Labopin M, Canaani J, Niittyvuopio R, Socié G, Kröger N, Itäla-Remes M, Yakoub-Agha I, Labussière-Wallet H, Gallego-Hernanz MP, Deconinck E, Chevallier P, Finke J, Esteve J, Mohty M. Cytogenetic risk score maintains its prognostic significance in AML patients with detectable measurable residual disease undergoing transplantation in remission: On behalf of the acute leukemia working party of the European society for blood and marrow transplantation. Am J Hematol 2020; 95:1135-1141. [PMID: 32530520 DOI: 10.1002/ajh.25905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/03/2020] [Accepted: 06/09/2020] [Indexed: 12/22/2022]
Abstract
While evidence for measurable residual disease (MRD) is a harbinger of inferior outcome in acute myeloid leukemia (AML) patients referred for allogeneic stem cell transplantation (allo-SCT), the exact clinical trajectory of specific patient subsets in this clinical setting is undefined. Using a recently published prognostic cytogenetic model (Canaani et al. Leukemia 2019) we evaluated whether this model applied also to studies of patients with positive MRD. The analysis comprised MRD+ patients in first complete remission undergoing allo-SCT from a matched sibling donor or unrelated donor. Seven hundred and seventy-five patients were evaluated with a median follow-up duration of 22 months. Cytogenetic risk score was favorable, intermediate/FLT3wt intermediate/FLT3-ITD3, and adverse in 15%, 28.3%, 37% and 19.7% of the patients, respectively. Favorable and intermediate/FLT3wt risk patients had 2-year leukemia-free survival rates of 78% and 61%, respectively, compared with only 50% and 37% for intermediate/FLT3-ITD3 and adverse risk patients, respectively (P < .001). In multivariate analysis adverse and intermediate/FLT3-ITD3 risk patients were more likely to experience disease relapse compared with favorable risk patients [hazard ratio (HR) = 3.9, 95% confidence interval (CI), 2.1-7.3; P < .001, and HR = 4.4, CI 95%, 2.4-7.8; P < .001, respectively]. The European society for blood and marrow transplantation cytogenetic risk score is a valuable adjunct for risk stratification of MRD+ AML patients.
Collapse
Affiliation(s)
- Arnon Nagler
- Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
- EBMT ALWP office, Saint Antoine Hospital, Paris, France
| | - Myriam Labopin
- Department of Haematology and EBMT Paris study office / CEREST-TC, Saint Antoine Hospital, Paris, France
| | - Jonathan Canaani
- Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Riitta Niittyvuopio
- HUCH Comprehensive Cancer Center, Stem Cell Transplantation Unit, Helsinki, Finland
| | - Gerard Socié
- Department of Hematology - BMT, Hopital St. Louis, Paris, France
| | - Nicolaus Kröger
- Bone Marrow Transplantation Centre, University Hospital Eppendorf, Hamburg, Germany
| | - Maija Itäla-Remes
- TD7 (Stem Cell Transplant Unit), Turku University Hospital, Turku, Finland
| | | | | | | | - Eric Deconinck
- Hopital Jean Minjoz, Service d'Hématologie, Besancon, France
| | | | - Jürgen Finke
- Department of Medicine -Hematology, Oncology, University of Freiburg, Freiburg, Germany
| | - Jordi Esteve
- Hematology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Mohamad Mohty
- Department of Haematology and EBMT Paris study office / CEREST-TC, Saint Antoine Hospital, INSERM UMR 938 and Université Pierre et Marie Curie, Paris, France
| |
Collapse
|
155
|
Bejanyan N, Zhang M, Bo-Subait K, Brunstein C, Wang H, Warlick ED, Giralt S, Nishihori T, Martino R, Passweg J, Dias A, Copelan E, Hale G, Gale RP, Solh M, Kharfan-Dabaja MA, Diaz MA, Ganguly S, Gore S, Verdonck LF, Hossain NM, Kekre N, Savani B, Byrne M, Kanakry C, Cairo MS, Ciurea S, Schouten HC, Bredeson C, Munker R, Lazarus H, Cahn JY, van Der Poel M, Rizzieri D, Yared JA, Freytes C, Cerny J, Aljurf M, Palmisiano ND, Pawarode A, Bacher VU, Grunwald MR, Nathan S, Wirk B, Hildebrandt GC, Seo S, Olsson RF, George B, de Lima M, Hourigan CS, Sandmaier BM, Litzow M, Kebriaei P, Saber W, Weisdorf D. Myeloablative Conditioning for Allogeneic Transplantation Results in Superior Disease-Free Survival for Acute Myelogenous Leukemia and Myelodysplastic Syndromes with Low/Intermediate but not High Disease Risk Index: A Center for International Blood and Marrow Transplant Research Study. Transplant Cell Ther 2020; 27:68.e1-68.e9. [PMID: 33010430 DOI: 10.1016/j.bbmt.2020.09.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/05/2020] [Accepted: 09/19/2020] [Indexed: 11/26/2022]
Abstract
Compared with reduced-intensity conditioning (RIC), myeloablative conditioning (MAC) is generally associated with lower relapse risk after allogeneic hematopoietic cell transplantation (HCT) for acute myelogenous leukemia (AML) and myelodysplastic syndromes (MDS). However, disease-specific risk factors in AML/MDS can further inform when MAC and RIC may yield differential outcomes. We analyzed HCT outcomes stratified by the Disease Risk Index (DRI) in 4387 adults (age 40 to 65 years) to identify the impact of conditioning intensity. In the low/intermediate-risk DRI cohort, RIC was associated with lower nonrelapse mortality (NRM) (hazard ratio [HR], .74; 95% confidence interval [CI], .62 to .88; P < .001) but significantly greater relapse risk (HR, 1.54; 95% CI, 1.35 to 1.76; P < .001) and thus inferior disease-free survival (DFS) (HR, 1.19; 95% CI, 1.07 to 1.33; P = .001). In the high/very high-risk DRI cohort, RIC was associated with marginally lower NRM (HR, .83; 95% CI, .68 to 1.00; P = .051) and significantly higher relapse risk (HR, 1.23; 95% CI, 1.08 to 1.41; P = .002), leading to similar DFS using either RIC or MAC. These data support MAC over RIC as the preferred conditioning intensity for patients with AML/MDS with low/intermediate-risk DRI, but with a similar benefit as RIC in high/very high-risk DRI. Novel MAC regimens with less toxicity could benefit all patients, but more potent antineoplastic approaches are needed for the high/very-high risk DRI group.
Collapse
Affiliation(s)
- Nelli Bejanyan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.
| | - Meijie Zhang
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Khalid Bo-Subait
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Claudio Brunstein
- Adult Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Hailin Wang
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Erica D Warlick
- Adult Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Sergio Giralt
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Rodrigo Martino
- Division of Clinical Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jakob Passweg
- Division of Hematology, Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Ajoy Dias
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Edward Copelan
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - Gregory Hale
- Department of Hematology/Oncology, Johns Hopkins All Children's Hospital, St Petersburg, Florida
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Melhem Solh
- Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, Georgia
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Miguel Angel Diaz
- Department of Hematology/Oncology, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Siddhartha Ganguly
- Division of Hematological Malignancy and Cellular Therapeutics, University of Kansas Health System, Kansas City, Kansas
| | - Steven Gore
- Section of Medical Oncology, Department of Internal Medicine, Yale New Haven Hospital, New Haven, Connecticut
| | - Leo F Verdonck
- Department of Hematology/Oncology, Isala Clinic, Zwolle, The Netherland
| | - Nasheed M Hossain
- Stem Cell Transplant Program, Division of Hematology/Oncology, Department of Medicine, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Natasha Kekre
- Blood & Marrow Transplant Program, Department of Medicine, Ottawa Hospital Ottawa, Ontario, Canada
| | - Bipin Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael Byrne
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christopher Kanakry
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mitchell S Cairo
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, New York Medical College, Valhalla, New York
| | - Stefan Ciurea
- Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Harry C Schouten
- Department of Hematology, Academische Ziekenhuis, Maastricht, The Netherlands
| | - Christopher Bredeson
- Blood & Marrow Transplant Program, Department of Medicine, Ottawa Hospital Ottawa, Ontario, Canada
| | - Reinhold Munker
- Division of Medical Oncology, Markey Cancer Center, University of Kentucky School of Medicine, Lexington, Kentucky
| | - Hillard Lazarus
- Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Jean-Yves Cahn
- Department of Hematology, CHU Grenoble Alpes, Grenoble, France
| | - Marjolein van Der Poel
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - David Rizzieri
- Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, North Carolina
| | - Jean A Yared
- Blood & Marrow Transplantation Program, Division of Hematology/Oncology, Department of Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Cesar Freytes
- Hematopoietic Stem Cell Transplant Program, Texas Transplant Institute, San Antonio, Texas
| | - Jan Cerny
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical Center, Worcester, Massachusetts
| | - Mahmoud Aljurf
- Department of Oncology, King Faisal Specialist Hospital Center & Research, Riyadh, Saudi Arabia
| | - Neil D Palmisiano
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Attaphol Pawarode
- Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Vera Ulrike Bacher
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - Sunita Nathan
- Section of Bone Marrow Transplantation and Cellular Therapy, Division of Hematology, Oncology and Cell Therapy, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | - Baldeep Wirk
- Bone Marrow Transplant Program, Penn State Cancer Institute, Hershey, Pennsylvania
| | - Gerhard C Hildebrandt
- Division of Medical Oncology, Markey Cancer Center, University of Kentucky School of Medicine, Lexington, Kentucky
| | - Sachiko Seo
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| | - Richard F Olsson
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Clinical Research Sormland, Uppsala University, Sweden
| | - Biju George
- Department of Haematology, Christian Medical College, Vellore, India
| | - Marcos de Lima
- Department of Medicine, Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Brenda M Sandmaier
- Division of Medical Oncology, University of Washington and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark Litzow
- Division of Hematology and Transplant Center, Mayo Clinic Rochester, Rochester, Minnesota
| | - Partow Kebriaei
- Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wael Saber
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daniel Weisdorf
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, 96
| |
Collapse
|
156
|
Mushtaq MU, Harrington AM, Chaudhary SG, Michaelis LC, Carlson KSB, Abedin S, Runass L, Callander NS, Fallon MJ, Juckett M, Hall AC, Hematti P, Mattison RJ, Atallah EL, Guru Murthy GS. Comparison of salvage chemotherapy regimens and prognostic significance of minimal residual disease in relapsed/refractory acute myeloid leukemia. Leuk Lymphoma 2020; 62:158-166. [PMID: 32951486 DOI: 10.1080/10428194.2020.1821009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We compared the outcomes of salvage chemotherapy in 146 patients with relapsed (57.5%) or refractory (42.5%) AML who received CLAG-M (51%), MEC (39%) or CLAG (10%). Minimal residual disease (MRD) was assessed by flow cytometry. Bivariate, Kaplan-Meier, and Cox regression analyses were conducted. Complete remission (CR) rate of 46% (CLAG-M 54% versus MEC/CLAG 40%, p = .045) was observed with MRD-negative CR of 33% (CLAG-M 39% versus MEC/CLAG 22%, p = .042). Median overall survival (OS) was 9.7 months; the longest OS occurred with CLAG-M (13.3, 95%CI 2.4-24.3) versus MEC (6.9, 95%CI 2.9-10.9) or CLAG (6.2, 95%CI 2.4-12.6) (p = .025). When adjusted for age, gender, relapsed/refractory AML, poor risk AML, MRD, chemotherapy and transplant, CLAG-M (HR 0.63, 95% CI 0.40-0.98, p = .042), MRD-negativity (HR 0.15, 95% CI 0.07-0.30, p < .001) and transplant (HR 0.22, 95% CI 0.13-0.39, p < .001) were associated with higher OS. Our findings confirm that CLAG-M is a reasonable salvage regimen for RR-AML followed by transplant.
Collapse
Affiliation(s)
- Muhammad Umair Mushtaq
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | | | - Sibgha Gull Chaudhary
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Laura C Michaelis
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Karen-Sue B Carlson
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sameem Abedin
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lyndsey Runass
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Natalie S Callander
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | | | - Mark Juckett
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Aric C Hall
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Peiman Hematti
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ryan J Mattison
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ehab L Atallah
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | | |
Collapse
|
157
|
Hernandez-Valladares M, Bruserud Ø, Selheim F. The Implementation of Mass Spectrometry-Based Proteomics Workflows in Clinical Routines of Acute Myeloid Leukemia: Applicability and Perspectives. Int J Mol Sci 2020; 21:ijms21186830. [PMID: 32957646 PMCID: PMC7556012 DOI: 10.3390/ijms21186830] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 02/08/2023] Open
Abstract
With the current reproducibility of proteome preparation workflows along with the speed and sensitivity of the mass spectrometers, the transition of the mass spectrometry (MS)-based proteomics technology from biomarker discovery to clinical implementation is under appraisal in the biomedicine community. Therefore, this technology might be implemented soon to detect well-known biomarkers in cancers and other diseases. Acute myeloid leukemia (AML) is an aggressive heterogeneous malignancy that requires intensive treatment to cure the patient. Leukemia relapse is still a major challenge even for patients who have favorable genetic abnormalities. MS-based proteomics could be of great help to both describe the proteome changes of individual patients and identify biomarkers that might encourage specific treatments or clinical strategies. Herein, we will review the advances and availability of the MS-based proteomics strategies that could already be used in clinical proteomics. However, the heterogeneity of complex diseases as AML requires consensus to recognize AML biomarkers and to establish MS-based workflows that allow their unbiased identification and quantification. Although our literature review appears promising towards the utilization of MS-based proteomics in clinical AML in a near future, major efforts are required to validate AML biomarkers and agree on clinically approved workflows.
Collapse
MESH Headings
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Computational Biology
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Mass Spectrometry/methods
- Prognosis
- Proteome/analysis
- Proteome/metabolism
- Proteomics/methods
- Robotics/instrumentation
- Robotics/methods
- Workflow
Collapse
Affiliation(s)
- Maria Hernandez-Valladares
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- The Proteomics Facility of the University of Bergen (PROBE), Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- Correspondence: (M.H.-V.); (Ø.B.); (F.S.); Tel.: +47-55586368 (M.H.-V.); +47-55972997 (Ø.B.); +47-55586368 (F.S.)
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Correspondence: (M.H.-V.); (Ø.B.); (F.S.); Tel.: +47-55586368 (M.H.-V.); +47-55972997 (Ø.B.); +47-55586368 (F.S.)
| | - Frode Selheim
- The Proteomics Facility of the University of Bergen (PROBE), Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- The Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- Correspondence: (M.H.-V.); (Ø.B.); (F.S.); Tel.: +47-55586368 (M.H.-V.); +47-55972997 (Ø.B.); +47-55586368 (F.S.)
| |
Collapse
|
158
|
Huang A, Chen Q, Fei Y, Wang Z, Ni X, Gao L, Chen L, Chen J, Zhang W, Yang J, Wang J, Hu X. Dynamic prediction of relapse in patients with acute leukemias after allogeneic transplantation: Joint model for minimal residual disease. Int J Lab Hematol 2020; 43:84-92. [PMID: 32881394 DOI: 10.1111/ijlh.13328] [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] [Received: 02/11/2020] [Revised: 07/21/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Relapse remains the leading cause of treatment failure after allogeneic hematopoietic stem cell transplantation (alloHSCT) in leukemia. Numerous investigations have demonstrated that minimal residual disease (MRD) before or after alloHSCT is prognostic of relapse risk. These MRD data were collected at specific checkpoints and could not dynamically predict the relapse risk after alloHSCT, which needs serial monitoring. METHODS In the present study, we retrospectively analyzed MRD measured with multi-parameter flow cytometry in 207 acute myeloid leukemia (AML) patients (acute promyelocytic leukemia excluded), and 124 acute B lymphoblastic leukemia (ALL) patients. A three-step method based on joint model was used to build a relapse risk prediction model. RESULTS The 3-year overall survival and relapse-free survival rates of the entire cohort were 67.1% ± 2.8% and 61.6% ± 2.8%, respectively. The model included disease status before alloHSCT, acute and chronic graft-versus-host disease, and serial MRD data. The time-dependent receiver operating characteristics was used to evaluate the ability of the model. It fitted well with actual incidence of relapse. The serial MRD data collected after alloHSCT had better discrimination capabilities for recurrence prediction with the area under the curve from 0.67 to 0.91 (AML: 0.66-0.89; ALL: 0.70-0.96). CONCLUSION The joint model was able to dynamically predict relapse-free probability after alloHSCT, which would be a useful tool to provide important information to guide decision-making in the clinic and facilitate the individualized therapy.
Collapse
Affiliation(s)
- Aijie Huang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Qi Chen
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Yang Fei
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Ziwei Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Xiong Ni
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Lei Gao
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Li Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Jie Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Weiping Zhang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Jianmin Yang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Jianmin Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Xiaoxia Hu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| |
Collapse
|
159
|
Morsink LM, Sandmaier BM, Othus M, Palmieri R, Granot N, Bezerra ED, Wood BL, Mielcarek M, Schoch G, Davis C, Flowers MED, Deeg HJ, Appelbaum FR, Storb R, Walter RB. Conditioning Intensity, Pre-Transplant Flow Cytometric Measurable Residual Disease, and Outcome in Adults with Acute Myeloid Leukemia Undergoing Allogeneic Hematopoietic Cell Transplantation. Cancers (Basel) 2020; 12:cancers12092339. [PMID: 32825022 PMCID: PMC7565021 DOI: 10.3390/cancers12092339] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
How conditioning intensity is related to outcomes of AML patients undergoing allografting in morphologic remission is an area of great ongoing interest. We studied 743 patients in morphologic remission and known pre-transplant measurable residual disease (MRD) status determined by multiparameter flow cytometry (MFC) who received a first allograft after myeloablative, reduced intensity, or nonmyeloablative conditioning (MAC, RIC, and NMA). Overall, relapse-free survival (RFS) and overall survival (OS) were longer after MAC than RIC or NMA conditioning, whereas relapse risks were not different. Among MRDpos patients, 3-year estimates of relapse risks and survival were similar across conditioning intensities. In contrast, among MRDneg patients, 3-year RFS and OS were longer for MAC (69% and 71%) than RIC (47% and 55%) and NMA conditioning (47% and 52%). Three-year relapse risks were lowest after MAC (18%) and highest after NMA conditioning (30%). Our data indicate an interaction between conditioning intensity, MFC-based pre-transplant MRD status, and outcome, with benefit of intensive conditioning primarily for patients transplanted in MRDneg remission. Differing from recent findings from other studies that indicated MAC is primarily beneficial for some or all patients with MRDpos pre-HCT status, our data suggest MAC should still be considered for MRDneg AML patients if tolerated.
Collapse
Affiliation(s)
- Linde M. Morsink
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
- Department of Hematology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Brenda M. Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Megan Othus
- Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
| | - Raffaele Palmieri
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
| | - Noa Granot
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
| | - Evandro D. Bezerra
- Department of Medicine, Residency Program, University of Washington, Seattle, WA 98195, USA;
| | - Brent L. Wood
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA;
| | - Marco Mielcarek
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Gary Schoch
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
| | - Chris Davis
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
| | - Mary E. D. Flowers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - H. Joachim Deeg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Frederick R. Appelbaum
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Rainer Storb
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (L.M.M.); (B.M.S.); (R.P.); (N.G.); (M.M.); (G.S.); (C.D.); (M.E.D.F.); (H.J.D.); (F.R.A.); (R.S.)
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA;
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
- Correspondence: ; Tel.: +1-206-667-3599
| |
Collapse
|
160
|
Nagler A, Baron F, Labopin M, Polge E, Esteve J, Bazarbachi A, Brissot E, Bug G, Ciceri F, Giebel S, Gilleece MH, Gorin NC, Lanza F, Peric Z, Ruggeri A, Sanz J, Savani BN, Schmid C, Shouval R, Spyridonidis A, Versluis J, Mohty M. Measurable residual disease (MRD) testing for acute leukemia in EBMT transplant centers: a survey on behalf of the ALWP of the EBMT. Bone Marrow Transplant 2020; 56:218-224. [PMID: 32724200 DOI: 10.1038/s41409-020-01005-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/07/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022]
Abstract
Detectable measurable residual disease (MRD) is a key prognostic factor in both acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) patients. Thus, we conducted a survey in EBMT transplant centers focusing on pre- and post-allo-HCT MRD. One hundred and six centers from 29 countries responded. One hundred had a formal strategy for routine MRD assessment, 91 for both ALL and AML. For ALL (n = 95), assessing MRD has been routine practice starting from 2010 (range, 1990-2019). Techniques used for MRD assessment consisted of PCR techniques alone (n = 27), multiparameter flow cytometry (MFC, n = 16), both techniques (n = 43), next-generation sequencing (NGS) + PCR (n = 2), or PCR + MFC + NGS (n = 7). The majority of centers assessed MRD every 2-3 months for 2 (range, 1-until relapse) years. For AML, assessing MRD was routine in 92 centers starting in 2010 (range 1990-2019). Assessment of MRD was by PCR (n = 23), MFC (n = 13), both PCR and MFC (n = 39), both PCR and NGS (n = 3), and by all three techniques (n = 14). The majority assesses MRD for AML every 2-3 months for 2 (range, 1-until relapse) years. This survey is the first step in the aim to include MRD status as a routine registry capture parameter in acute leukemia.
Collapse
Affiliation(s)
- Arnon Nagler
- Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Tel Aviv, Israel. .,EBMT ALWP Office, Saint Antoine Hospital, Paris, France.
| | - Frédéric Baron
- Department of Hematology, University of Liège, Liège, Belgium
| | - Myriam Labopin
- EBMT Paris Study Office/CEREST-TC, Paris, France.,Department of Haematology, Saint Antoine Hospital, Paris, France.,INSERM UMR 938, Paris, France.,Sorbonne University, Paris, France
| | | | - Jordi Esteve
- Hematology Department, Hospital Clinic, Barcelona, Spain
| | - Ali Bazarbachi
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Eolia Brissot
- Department of Haematology, Saint Antoine Hospital, Paris, France.,INSERM UMR 938, Paris, France.,Sorbonne University, Paris, France
| | - Gesine Bug
- Medizinische Klinik II, Hämatologie, Medizinische Onkologie, Goethe-Universitaet, Frankfurt, Germany
| | - Fabio Ciceri
- Ospedale San Raffaele S.r.l., Haematology and BMT, Milan, Italy
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie Institute, Oncology Center, Gliwice, Poland
| | - Maria H Gilleece
- Yorkshire Blood and Marrow Transplant Programme, Haematology Department, St James's Institute of Oncology, Leeds, UK
| | | | | | - Zinaida Peric
- Bone Marrow Transplant Unit, University Hospital Center Rebro, Zagreb, Croatia
| | | | - Jaime Sanz
- Hematology Department, University Hospital La Fe, Valencia, Spain
| | - Bipin N Savani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Roni Shouval
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel
| | | | - Jurjen Versluis
- Erasmus University Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Mohamad Mohty
- EBMT Paris Study Office/CEREST-TC, Paris, France.,Department of Haematology, Saint Antoine Hospital, Paris, France.,INSERM UMR 938, Paris, France.,Sorbonne University, Paris, France
| |
Collapse
|
161
|
Recent advances in allogeneic hematopoietic cell transplantation for acute myeloid leukemia. Curr Opin Hematol 2020; 27:115-121. [PMID: 31913152 DOI: 10.1097/moh.0000000000000572] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Allogeneic hematopoietic cell transplantation (HCT), with associated graft-versus-leukemia effects, remains the best postremission strategy for patients with intermediate or high-risk acute myeloid leukemia (AML), with a curative potential. Here, we highlight recent advances in allogeneic HCT that broadened access, refined prognostication, and improved outcomes of AML patients undergoing this procedure. RECENT FINDINGS Eligibility for allogeneic HCT continued to expand to AML patients older than 60 years, as well as to patients lacking human leukocyte antigen (HLA)-matched donors with the advent of alternative donor sources, such as umbilical cord blood and HLA-haploidentical transplantation. Molecular profiling of AML has redefined prognostication for patients in specific AML genomic subgroups undergoing allogeneic HCT and has served as a new strategy for measuring minimal residual disease before and after allogeneic HCT. Using high intensity conditioning regimens has emerged as a potential strategy to reduce risk of relapse and improve overall survival, especially in patients with minimal residual disease prior to allogeneic HCT. SUMMARY As access to allogeneic HCT continues to improve, also, with more refined prognostic strategies, the field continues to move to optimize transplantation approaches by decreasing the risk of relapse and minimizing transplant-related complications.
Collapse
|
162
|
Hourigan CS, Dillon LW, Gui G, Logan BR, Fei M, Ghannam J, Li Y, Licon A, Alyea EP, Bashey A, Deeg HJ, Devine SM, Fernandez HF, Giralt S, Hamadani M, Howard A, Maziarz RT, Porter DL, Scott BL, Warlick ED, Pasquini MC, Horwitz ME. Impact of Conditioning Intensity of Allogeneic Transplantation for Acute Myeloid Leukemia With Genomic Evidence of Residual Disease. J Clin Oncol 2020; 38:1273-1283. [PMID: 31860405 PMCID: PMC7164487 DOI: 10.1200/jco.19.03011] [Citation(s) in RCA: 304] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Patients with acute myeloid leukemia (AML) in remission remain at risk for relapse even after allogeneic hematopoietic cell transplantation (alloHCT). AML measurable residual disease (MRD) status before alloHCT has been shown to be prognostic. Whether modulation of the intensity of the alloHCT conditioning regimen in patients with AML who test positive for MRD can prevent relapse and improve survival is unknown. METHODS Ultra-deep, error-corrected sequencing for 13 commonly mutated genes in AML was performed on preconditioning blood from patients treated in a phase III clinical trial that randomly assigned adult patients with myeloid malignancy in morphologic complete remission to myeloablative conditioning (MAC) or reduced-intensity conditioning (RIC). RESULTS No mutations were detected in 32% of MAC and 37% of RIC recipients; these groups had similar survival (3-year overall survival [OS], 56% v 63%; P = .96). In patients with a detectable mutation (next-generation sequencing [NGS] positive), relapse (3-year cumulative incidence, 19% v 67%; P < .001) and survival (3-year OS, 61% v 43%; P = .02) was significantly different between the MAC and RIC arms, respectively. In multivariable analysis for NGS-positive patients, adjusting for disease risk and donor group, RIC was significantly associated with increased relapse (hazard ratio [HR], 6.38; 95% CI, 3.37 to 12.10; P < .001), decreased relapse-free survival (HR, 2.94; 95% CI, 1.84 to 4.69; P < .001), and decreased OS (HR, 1.97; 95% CI, 1.17 to 3.30; P = .01) compared with MAC. Models of AML MRD also showed benefit for MAC over RIC for those who tested positive. CONCLUSION This study provides evidence that MAC rather than RIC in patients with AML with genomic evidence of MRD before alloHCT can result in improved survival.
Collapse
MESH Headings
- Adult
- Aged
- Circulating Tumor DNA/blood
- Circulating Tumor DNA/genetics
- Clinical Trials, Phase III as Topic
- Female
- Hematopoietic Stem Cell Transplantation/methods
- High-Throughput Nucleotide Sequencing
- Humans
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Mutation
- Neoplasm, Residual
- Prognosis
- Randomized Controlled Trials as Topic
- Transplantation Conditioning/methods
- Transplantation, Homologous
- Young Adult
Collapse
Affiliation(s)
| | - Laura W. Dillon
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Gege Gui
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | | | - Jack Ghannam
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Yuesheng Li
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | | | - Asad Bashey
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA
| | | | - Steven M. Devine
- National Marrow Donor Program and Center for International Blood and Marrow Transplant Research, Minneapolis, MN
| | | | - Sergio Giralt
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Alan Howard
- National Marrow Donor Program and Center for International Blood and Marrow Transplant Research, Minneapolis, MN
| | - Richard T. Maziarz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | | | | | | | | | | |
Collapse
|
163
|
Freeman S, Craddock C. Less Is Not Necessarily More: Toward a Rational Selection of the Conditioning Regimen in Acute Myeloid Leukemia. J Clin Oncol 2020; 38:1249-1251. [DOI: 10.1200/jco.19.03161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Sylvie Freeman
- Institute of Infection and Immunity University of Birmingham, Edgbaston, Birmingham, United Kingdom
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- Cancer Research UK Cancer Trials Unit, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
164
|
Marconi G, Talami A, Abbenante MC, Sartor C, Parisi S, Nanni J, Bertamini L, Ragaini S, Olivi M, de Polo S, Cristiano G, Fontana MC, Bochicchio MT, Ottaviani E, Arpinati M, Sessa M, Baldazzi C, Caso L, Testoni N, Baccarani M, Bonifazi F, Martinelli G, Paolini S, Cavo M, Papayannidis C, Curti A. MEC (mitoxantrone, etoposide, and cytarabine) induces complete remission and is an effective bridge to transplant in acute myeloid leukemia. Eur J Haematol 2020; 105:47-55. [DOI: 10.1111/ejh.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Giovanni Marconi
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Annalisa Talami
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Maria Chiara Abbenante
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
- Department of Haematology and Stem Cell Transplantation Unit IRCCS “Casa Sollievo della Sofferenza” Hospital San Giovanni Rotondo Italy
| | - Chiara Sartor
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Sarah Parisi
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Jacopo Nanni
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Luca Bertamini
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
- Division of Hematology AOU Città della Salute e della Scienza di Torino Torino Italy
| | - Simone Ragaini
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Matteo Olivi
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Stefano de Polo
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Gianluca Cristiano
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Maria Chiara Fontana
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Maria Teresa Bochicchio
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS Meldola Italy
| | - Emanuela Ottaviani
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Mario Arpinati
- Department of Hematology and Oncology Institute of Hematology L. e A. Seràgnoli Azienda Ospedaliero‐Universitaria S. Orsola Malpighi Bologna Italy
| | - Mariarosaria Sessa
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Carmen Baldazzi
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Lucia Caso
- Department of Hematology and Oncology Institute of Hematology L. e A. Seràgnoli Azienda Ospedaliero‐Universitaria S. Orsola Malpighi Bologna Italy
| | - Nicoletta Testoni
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Michele Baccarani
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Francesca Bonifazi
- Department of Hematology and Oncology Institute of Hematology L. e A. Seràgnoli Azienda Ospedaliero‐Universitaria S. Orsola Malpighi Bologna Italy
| | - Giovanni Martinelli
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS Meldola Italy
| | - Stefania Paolini
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Michele Cavo
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Cristina Papayannidis
- Department of Experimental, Diagnostic and Specialty Medicine Institute of Hematology "L. and A. Seràgnoli" University of Bologna Bologna Italy
| | - Antonio Curti
- Department of Hematology and Oncology Institute of Hematology L. e A. Seràgnoli Azienda Ospedaliero‐Universitaria S. Orsola Malpighi Bologna Italy
| |
Collapse
|
165
|
Gavillet M, Carr Klappert J, Spertini O, Blum S. Acute leukemia in the time of COVID-19. Leuk Res 2020; 92:106353. [PMID: 32251934 PMCID: PMC7138175 DOI: 10.1016/j.leukres.2020.106353] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Mathilde Gavillet
- Service and Central Laboratory of Hematology, Department of Oncology and Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV), Lausanne, Switzerland.
| | - Jeanette Carr Klappert
- Service and Central Laboratory of Hematology, Department of Oncology and Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Olivier Spertini
- Service and Central Laboratory of Hematology, Department of Oncology and Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Sabine Blum
- Service and Central Laboratory of Hematology, Department of Oncology and Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
166
|
Venditti A, Gale RP, Buccisano F, Ossenkoppele G. Should persons with acute myeloid leukemia (AML) in 1st histological complete remission who are measurable residual disease (MRD) test positive receive an allotransplant? Leukemia 2020; 34:963-965. [PMID: 32132654 DOI: 10.1038/s41375-020-0780-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/01/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Adriano Venditti
- Department of Biomedicine and Prevention at University "Tor Vergata", Fondazione Policlinico Tor Vergata, Rome, Italy.
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Francesco Buccisano
- Department of Biomedicine and Prevention at University "Tor Vergata", Fondazione Policlinico Tor Vergata, Rome, Italy
| | - Gert Ossenkoppele
- Department of Hematology, VU University Medical Centre Amsterdam, Amsterdam, Noord-Holland, The Netherlands
| |
Collapse
|
167
|
Dillon R, Hills R, Freeman S, Potter N, Jovanovic J, Ivey A, Kanda AS, Runglall M, Foot N, Valganon M, Khwaja A, Cavenagh J, Smith M, Ommen HB, Overgaard UM, Dennis M, Knapper S, Kaur H, Taussig D, Mehta P, Raj K, Novitzky-Basso I, Nikolousis E, Danby R, Krishnamurthy P, Hill K, Finnegan D, Alimam S, Hurst E, Johnson P, Khan A, Salim R, Craddock C, Spearing R, Gilkes A, Gale R, Burnett A, Russell NH, Grimwade D. Molecular MRD status and outcome after transplantation in NPM1-mutated AML. Blood 2020; 135:680-688. [PMID: 31932839 PMCID: PMC7059484 DOI: 10.1182/blood.2019002959] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
Relapse remains the most common cause of treatment failure for patients with acute myeloid leukemia (AML) who undergo allogeneic stem cell transplantation (alloSCT), and carries a grave prognosis. Multiple studies have identified the presence of measurable residual disease (MRD) assessed by flow cytometry before alloSCT as a strong predictor of relapse, but it is not clear how these findings apply to patients who test positive in molecular MRD assays, which have far greater sensitivity. We analyzed pretransplant blood and bone marrow samples by reverse-transcription polymerase chain reaction in 107 patients with NPM1-mutant AML enrolled in the UK National Cancer Research Institute AML17 study. After a median follow-up of 4.9 years, patients with negative, low (<200 copies per 105ABL in the peripheral blood and <1000 copies in the bone marrow aspirate), and high levels of MRD had an estimated 2-year overall survival (2y-OS) of 83%, 63%, and 13%, respectively (P < .0001). Focusing on patients with low-level MRD before alloSCT, those with FLT3 internal tandem duplications(ITDs) had significantly poorer outcome (hazard ratio [HR], 6.14; P = .01). Combining these variables was highly prognostic, dividing patients into 2 groups with 2y-OS of 17% and 82% (HR, 13.2; P < .0001). T-depletion was associated with significantly reduced survival both in the entire cohort (2y-OS, 56% vs 96%; HR, 3.24; P = .0005) and in MRD-positive patients (2y-OS, 34% vs 100%; HR, 3.78; P = .003), but there was no significant effect of either conditioning regimen or donor source on outcome. Registered at ISRCTN (http://www.isrctn.com/ISRCTN55675535).
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Female
- Hematopoietic Stem Cell Transplantation/mortality
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/mortality
- Neoplasm, Residual/diagnosis
- Neoplasm, Residual/genetics
- Nuclear Proteins/genetics
- Nucleophosmin
- Recurrence
- Young Adult
Collapse
Affiliation(s)
- Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Cancer Genetics Service, Viapath, Guy's Hospital, London, United Kingdom
- Department of Haematology, Guy's Hospital, London, United Kingdom
| | - Robert Hills
- Nuffield Department of Population Health, University of Oxford, United Kingdom
| | - Sylvie Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Nicola Potter
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Cancer Genetics Service, Viapath, Guy's Hospital, London, United Kingdom
| | - Jelena Jovanovic
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Adam Ivey
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Anju Shankar Kanda
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Manohursingh Runglall
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Nicola Foot
- Cancer Genetics Service, Viapath, Guy's Hospital, London, United Kingdom
| | - Mikel Valganon
- Cancer Genetics Service, Viapath, Guy's Hospital, London, United Kingdom
| | - Asim Khwaja
- Department of Haematology, University College, London, United Kingdom
| | | | | | | | | | - Mike Dennis
- Christie Hospital, Manchester, United Kingdom
| | - Steven Knapper
- Department of Haematology, Cardiff University, Cardiff, United Kingdom
| | - Harpreet Kaur
- Royal Hallamshire Hospital, Sheffield, United Kingdom
| | | | - Priyanka Mehta
- Bristol Haematology and Oncology Centre, Bristol, United Kingdom
| | - Kavita Raj
- Department of Haematology, Guy's Hospital, London, United Kingdom
| | | | | | | | | | - Kate Hill
- University Hospital, Southampton, United Kingdom
| | | | - Samah Alimam
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Department of Haematology, Guy's Hospital, London, United Kingdom
| | - Erin Hurst
- Royal Victoria Infirmary, Newcastle, United Kingdom
| | | | - Anjum Khan
- St James' Hospital, Leeds, United Kingdom
| | - Rahuman Salim
- Clatterbridge Cancer Centre, Liverpool, United Kingdom
| | | | | | - Amanda Gilkes
- Department of Haematology, Cardiff University, Cardiff, United Kingdom
| | - Rosemary Gale
- Department of Haematology, University College, London, United Kingdom
| | - Alan Burnett
- Blackwaterfoot, Isle of Arran, United Kingdom; and
| | - Nigel H Russell
- Department of Haematology, Guy's Hospital, London, United Kingdom
- Nottingham University Hospital, Nottingham, United Kingdom
| | - David Grimwade
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Department of Haematology, Guy's Hospital, London, United Kingdom
| |
Collapse
|
168
|
Abstract
OPINION STATEMENT The expanding availability of minimal or more precisely measurable residual disease (MRD) assessment in acute myeloid leukemia (AML) with its possible implications for therapeutic decisions is of high interest to clinicians treating AML patients. A variety of mostly retrospective studies have shown that AML patients with a positive MRD test, assessed by different techniques at defined cutoffs and time-points, are at significantly higher risk of relapse and experience shorter overall survival compared to MRD-negative patients. How this valuable information may be adapted in the daily routine of patients' treatment to distinguish individuals who need more aggressive therapy from the ones who can be spared additional therapy to avoid treatment-related toxicities is still being investigated. With the exception of MRD analyses in acute promyelocitic leukemia (APL), the clinical implications of MRD tests for the individual AML patient are still mostly unknown. We currently lack hard evidence that MRD-based therapy modulation during treatment or pre-emptive intervention in MRD-positive patients after therapy would improve outcomes in non-APL AML patients. These questions will be evaluated in prospective randomized clinical trials. Today, however, some conclusions with regard to MRD assessment in AML can be drawn from the published data and are reviewed in this article.
Collapse
|
169
|
Impact of pretransplant measurable residual disease on the outcome of allogeneic hematopoietic cell transplantation in adult monosomal karyotype AML. Leukemia 2020; 34:1577-1587. [PMID: 31974434 PMCID: PMC7272254 DOI: 10.1038/s41375-020-0717-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/05/2019] [Accepted: 01/14/2020] [Indexed: 12/28/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is often unsuccessful for monosomal karyotype (MK) acute myeloid leukemia (AML). To what degree failures are associated with pre-transplant measurable residual disease (MRD) – a dominant adverse risk factor – is unknown. We therefore studied 606 adults with intermediate- or adverse-risk AML in morphologic remission who underwent allogeneic HCT between 4/2006 and 1/2019. Sixty-eight (11%) patients had MK AML, the majority of whom with complex cytogenetics. Before HCT, MK AML patients more often tested MRDpos by multiparameter flow cytometry (49% vs. 18%; P<0.001) and more likely had persistent cytogenetic abnormalities (44% vs. 13%; P<0.001) than non-MK AML patients. Three-year relapse/overall survival estimates were 46%/43% and 72%/15% for MRDneg and MRDpos MK AML patients, respectively, contrasted to 20%/64% and 64%/38% for MRDneg and MRDpos non-MK AML patients, respectively. After multivariable adjustment, MRDpos remission status but not MK remained statistically significantly associated with shorter survival and higher relapse risk. Similar results were obtained in several patient subsets. In summary, while our study confirms higher relapse rates and shorter survival for MK-AML compared to non-MK AML patients, these outcomes are largely accounted for by the presence of other adverse prognostic factors, in particular higher likelihood of pre-HCT MRD.
Collapse
|
170
|
Dholaria B, Savani BN, Labopin M, Luznik L, Ruggeri A, Mielke S, Al Malki MM, Kongtim P, Fuchs E, Huang XJ, Locatelli F, Aversa F, Castagna L, Bacigalupo A, Martelli M, Blaise D, Ben Soussan P, Arnault Y, Handgretinger R, Roy DC, O'Donnell P, Bashey A, Solomon S, Romee R, Lewalle P, Gayoso J, Maschan M, Lazarus HM, Ballen K, Giebel S, Baron F, Ciceri F, Esteve J, Gorin NC, Spyridonidis A, Schmid C, Ciurea SO, Nagler A, Mohty M. Clinical applications of donor lymphocyte infusion from an HLA-haploidentical donor: consensus recommendations from the Acute Leukemia Working Party of the EBMT. Haematologica 2020; 105:47-58. [PMID: 31537691 PMCID: PMC6939532 DOI: 10.3324/haematol.2019.219790] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 09/19/2019] [Indexed: 01/30/2023] Open
Abstract
Donor lymphocyte infusion has been used in the management of relapsed hematologic malignancies after allogeneic hematopoietic cell transplantation. It can eradicate minimal residual disease or be used to rescue a hematologic relapse, being able to induce durable remissions in a subset of patients. With the increased use of haploidentical hematopoietic cell transplantation, there is renewed interest in the use of donor lymphocytes to either treat or prevent disease relapse post transplant. Published retrospective and small prospective studies have shown encouraging results with therapeutic donor lymphocyte infusion in different haploidentical transplantation platforms. In this consensus paper, finalized on behalf of the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation, we summarize the available evidence on the use of donor lymphocyte infusion from haploidentical donor, and provide recommendations on its therapeutic, pre-emptive and prophylactic use in clinical practice.
Collapse
Affiliation(s)
- Bhagirathbhai Dholaria
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bipin N Savani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Myriam Labopin
- Department of Haematology and EBMT Paris study office / CEREST-TC, Saint Antoine Hospital, Paris, France
| | - Leo Luznik
- Department of Oncology Hematologic Malignancies, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Annalisa Ruggeri
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Stephan Mielke
- Department of Laboratory Medicine, CAST, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Monzr M Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Piyanuch Kongtim
- Stem Cell Transplant and Cellular Therapy, Thammasat University, Pathumthani, Thailand
| | - Ephraim Fuchs
- Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing China
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Sapienza, University of Rome, Italy
| | - Franco Aversa
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Andrea Bacigalupo
- Fondazione Policlinico Universitario Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Didier Blaise
- Department of Hematology, Institut Paoli Calmettes, Marseille France
| | - Patrick Ben Soussan
- Department of Clinical Psychology, Paoli-Calmettes Institute, Marseille, France
| | - Yolande Arnault
- Institut Paoli-Calmette, département de psychologie clinique, Marseille, France
| | - Rupert Handgretinger
- Department of Hematology and Oncology, University Children's Hospital Tübingen, Tübingen Germany
| | - Denis-Claude Roy
- Division of Hematology and Medical Oncology, Hospital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Paul O'Donnell
- Hematology-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Asad Bashey
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA, USA
| | - Scott Solomon
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA, USA
| | - Rizwan Romee
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Philippe Lewalle
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Jorge Gayoso
- HGU Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Michael Maschan
- Oncology and immunology, Dmitriy Rogachev National Medical Center of pediatric hematology, Moscow, Russia
| | - Hillard M Lazarus
- Adult Hematologic Malignancies & Stem Cell Transplant Section, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Karen Ballen
- Division of hematology/oncology, University of Virginia Health System, Charlottesville, VA, USA
| | - Sebastian Giebel
- Dept. of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Frederic Baron
- Laboratory of Hematology, University of Liège, Liège, Belgium
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milano Italy
| | - Jordi Esteve
- Hematology department, Hospital Clínic de Barcelona, Barcelona Spain
| | - Norbert-Claude Gorin
- Service d'hématologie et thérapie cellulaire Centre international greffes APHP-EBMT-INCa Hospital, Saint Antoine Hospital, Paris France
| | - Alexandros Spyridonidis
- Bone Marrow Transplantation Unit and CBMDP Donor Center, University Hospital of Patras, Patras, Greece
| | - Christoph Schmid
- Department of Hematology and Oncology, Klinikum Augsburg, Augsburg, Germany
| | - Stefan O Ciurea
- Stem Cell Transplant and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arnon Nagler
- Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel and EBMT ALWP office, Saint Antoine Hospital, Paris, France
| | - Mohamad Mohty
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, AP-HP, Sorobonne University, and INSERM UMRs 938, Paris, France
| |
Collapse
|
171
|
Loke J, Malladi R, Moss P, Craddock C. The role of allogeneic stem cell transplantation in the management of acute myeloid leukaemia: a triumph of hope and experience. Br J Haematol 2020; 188:129-146. [PMID: 31823351 PMCID: PMC6972492 DOI: 10.1111/bjh.16355] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Acute myeloid leukaemia (AML) is the commonest indication for allogeneic stem cell transplantation (allo-SCT) worldwide. The accumulated experience of allografting in AML over the last four decades has provided critical insights into both the contribution of the conditioning regimen and the graft-versus-leukaemia effect to the curative potential of the most common form of immunotherapy utilised in standard clinical practice. Coupled with advances in donor availability and transplant technologies, this has resulted in allo-SCT becoming an important treatment modality for the majority of adults with high-risk AML. At the same time, advances in genomic classification, coupled with progress in the accurate quantification of measurable residual disease, have increased the precision with which allo-mandatory patients can be identified, whilst simultaneously permitting accurate identification of those patients who can be spared the toxicity of an allograft. Despite this progress, disease recurrence still remains a major cause of transplant failure and AML has served as a paradigm for the development of strategies to reduce the risk of relapse - notably the novel concept of post-transplant maintenance, utilising pharmacological or cellular therapies.
Collapse
Affiliation(s)
- Justin Loke
- Centre for Clinical HaematologyQueen Elizabeth HospitalBirminghamUK
- University of BirminghamBirminghamUK
| | - Ram Malladi
- Centre for Clinical HaematologyQueen Elizabeth HospitalBirminghamUK
- University of BirminghamBirminghamUK
| | - Paul Moss
- Centre for Clinical HaematologyQueen Elizabeth HospitalBirminghamUK
- University of BirminghamBirminghamUK
| | - Charles Craddock
- Centre for Clinical HaematologyQueen Elizabeth HospitalBirminghamUK
- University of BirminghamBirminghamUK
| |
Collapse
|
172
|
Song K, Khan F. Cancer rehabilitation during the COVID-19 pandemic: An overview of special considerations. THE JOURNAL OF THE INTERNATIONAL SOCIETY OF PHYSICAL AND REHABILITATION MEDICINE 2020. [DOI: 10.4103/jisprm.jisprm_10_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
173
|
Kongtim P, Hasan O, Perez JMR, Varma A, Wang SA, Patel KP, Chen J, Rondon G, Srour S, Bashir Q, Qazilbash M, Mehta R, Shpall EJ, Alousi A, Khouri I, Kebriaei P, Popat U, Champlin RR, Ciurea SO. Novel Disease Risk Model for Patients with Acute Myeloid Leukemia Receiving Allogeneic Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2020; 26:197-203. [PMID: 31518645 DOI: 10.1016/j.bbmt.2019.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/01/2019] [Accepted: 09/04/2019] [Indexed: 02/04/2023]
Abstract
Molecular data and minimal residual disease (MRD) have been shown to influence outcomes in acute myeloid leukemia (AML) patients undergoing allogeneic hematopoietic cell transplantation (AHCT). Here we developed and validated a novel AML-specific disease risk group (AML-DRG) and revised our previously developed hematopoietic cell transplant-composite risk (HCT-CR) model by incorporating molecular data and MRD status to predict outcomes of patients with AML. The study included 1414 consecutively treated adult AML patients who received a first AHCT. Patients were randomly assigned into training (n = 944) and validation (n = 470) sets. To develop the AML-DRG model, the coefficient of all significant AML-related variables in multivariable Cox regression analysis in a training dataset was converted into scores, whereas the AML-HCT-CR was the sum of disease-related factors assessed by the AML-DRG model with the addition of weighted scores from patient-related factors. The AML-DRG was developed by assigning the following scores: 1 point to secondary AML, 1 point to the European LeukaemiaNet adverse genetic risk, 2 points to complete remission with MRD positive/unknown, and 4 points to active disease. These scores were used to generate 3 risk groups of the AML-DRG with significantly different overall survivals. By adding the score for significant patient-related factors (HCT-specific comorbidity index/age), we created 4 risk groups of AML-HCT-CR with distinct survival outcomes. Both the AML-DRG and AML-HCT-CR provided significantly better discriminative capacity compared with the disease risk index, European LeukaemiaNet genetic risk model, and cytogenetic risk model. Prognostic models incorporating molecular data and MRD status allow better stratification and improved survival estimates of AML patients post-transplant.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Disease-Free Survival
- Female
- Hematopoietic Stem Cell Transplantation
- Humans
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Models, Biological
- Neoplasm, Residual
- Risk Assessment
- Survival Rate
- Transplantation Conditioning
Collapse
Affiliation(s)
- Piyanuch Kongtim
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas; Center of Excellence in Applied Epidemiology, Thammasat University, Pathumthani, Thailand; Department of Medicine Thammasat University, Pathumthani, Thailand
| | - Omar Hasan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Miguel Ramos Perez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ankur Varma
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julianne Chen
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Muzaffar Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rohtesh Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Issa Khouri
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard R Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan O Ciurea
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
174
|
Andreani G, Cilloni D. Strategies for minimal residual disease detection: current perspectives. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2019; 9:1-8. [PMID: 31807111 PMCID: PMC6855617 DOI: 10.2147/blctt.s172693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/13/2018] [Indexed: 11/25/2022]
Abstract
Currently, the post-remission treatment in acute leukemia is based on the genetic profile of leukemic cells at diagnosis (ie, FLT3 ITD positivity) and on the level of measurable residual disease (MRD) after induction and consolidation chemotherapy. Two methods are currently preferred for MRD evaluation in many centers: multiparameter flow cytometry and real-time quantitative PCR. Additional methods such as next-generation sequencing and digital PCR are under investigation, in an attempt to increase the sensitivity and thus allowing the detection of small clones. Many studies suggest that MRD positivity after chemotherapy is associated with negative prognosis, and the reappearance of MRD during follow-up allows impending relapse to be identified and consequently enables early intervention. Finally, MRD positivity before hematopoietic stem cell transplantation is predictive of the outcome. Although the significance of MRD in acute leukemia has been widely explored, the assessment of molecular MRD is not yet a routine practice. In this review, we describe the significance of MRD in different settings and the main markers and methods used for MRD detection.
Collapse
Affiliation(s)
- Giacomo Andreani
- Department of Clinical and Biological Sciences, University of Turin, Turin,Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Turin,Italy
| |
Collapse
|
175
|
Cloos J, Ossenkoppele GJ, Dillon R. Minimal residual disease and stem cell transplantation outcomes. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:617-625. [PMID: 31808862 PMCID: PMC6913494 DOI: 10.1182/hematology.2019000006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Risk classification and tailoring of treatment are essential for improving outcome for patients with acute myeloid leukemia or high-risk myelodysplastic syndrome. Both patient and leukemia-specific characteristics assessed using morphology, cytogenetics, molecular biology, and multicolor flow cytometry are relevant at diagnosis and during induction, consolidation, and maintenance phases of the treatment. In particular, minimal residual disease (MRD) during therapy has potential as a prognostic factor of outcome, determination of response to therapy, and direction of targeted therapy. MRD can be determined by cell surface markers using multicolor flow cytometry, whereas leukemia-specific translocations and mutations are measured using polymerase chain reaction-based techniques and recently using next-generation sequencing. All these methods of MRD detection have their (dis)advantages, and all need to be standardized, prospectively validated, and improved to be used for uniform clinical decision making and a potential surrogate end point for clinical trials testing novel treatment strategies. Important issues to be solved are time point of MRD measurement and threshold for MRD positivity. MRD is used for stem cell transplantation (SCT) selection in the large subgroup of patients with an intermediate risk profile. Patients who are MRD positive will benefit from allo-SCT. However, MRD-negative patients have a better chance of survival after SCT. Therefore, it is debated whether MRD-positive patients should be extensively treated to become MRD negative before SCT. Either way, accurate monitoring of potential residual or upcoming disease is mandatory. Tailoring therapy according to MRD monitoring may be the most successful way to provide appropriate specifically targeted, personalized treatment.
Collapse
Affiliation(s)
- Jacqueline Cloos
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, VUMC, Amsterdam, The Netherlands; and
| | - Gert J Ossenkoppele
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, VUMC, Amsterdam, The Netherlands; and
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| |
Collapse
|
176
|
Freeman SD, Hourigan CS. MRD evaluation of AML in clinical practice: are we there yet? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:557-569. [PMID: 31808906 PMCID: PMC6913462 DOI: 10.1182/hematology.2019000060] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
MRD technologies increase our ability to measure response in acute myeloid leukemia (AML) beyond the limitations of morphology. When applied in clinical trials, molecular and immunophenotypic MRD assays have improved prognostic precision, providing a strong rationale for their use to guide treatment, as well as to measure its effectiveness. Initiatives such as those from the European Leukemia Network now provide a collaborative knowledge-based framework for selection and implementation of MRD assays most appropriate for defined genetic subgroups. For patients with mutated-NPM1 AML, quantitative polymerase chain reaction (qPCR) monitoring of mutated-NPM1 transcripts postinduction and sequentially after treatment has emerged as a highly sensitive and specific tool to predict relapse and potential benefit from allogeneic transplant. Flow cytometric MRD after induction is prognostic across genetic risk groups and can identify those patients in the wild-type NPM1 intermediate AML subgroup with a very high risk for relapse. In parallel with these data, advances in genetic profiling have extended understanding of the etiology and the complex dynamic clonal nature of AML, as well as created the opportunity for MRD monitoring using next-generation sequencing (NGS). NGS AML MRD detection can stratify outcomes and has potential utility in the peri-allogeneic transplant setting. However, there remain challenges inherent in the NGS approach of multiplex quantification of mutations to track AML MRD. Although further development of this methodology, together with orthogonal testing, will clarify its relevance for routine clinical use, particularly for patients lacking a qPCR genetic target, established validated MRD assays can already provide information to direct clinical practice.
Collapse
Affiliation(s)
- Sylvie D Freeman
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; and
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
177
|
Ghannam J, Dillon LW, Hourigan CS. Next-generation sequencing for measurable residual disease detection in acute myeloid leukaemia. Br J Haematol 2019; 188:77-85. [PMID: 31804716 DOI: 10.1111/bjh.16362] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukaemia (AML) is a blood cancer characterized by acquired genetic mutations. There is great interest in accurately establishing measurable residual disease (MRD) burden in AML patients in remission after treatment but at risk of relapse. However, inter- and intrapatient genetic diversity means that, unlike in the chronic myeloid and acute promyelocytic leukaemias, no single genetic abnormality is pathognomonic for all cases of AML MRD. Next-generation sequencing offers the opportunity to test broadly and deeply for potential genetic evidence of residual AML, and while not currently accepted for such use clinically, is likely to be increasingly used for AML MRD testing in the future.
Collapse
Affiliation(s)
- Jack Ghannam
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Laura W Dillon
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
178
|
Srour SA, Saliba RM, Bittencourt MCB, Perez JMR, Kongtim P, Alousi A, Al‐Atrash G, Olson A, Betul O, Mehta R, Popat U, Hosing C, Bashir Q, Khouri I, Kebriaei P, Masarova L, Short N, Jabbour E, Daver N, Konopleva M, Ravandi F, Kantarjian H, Champlin RE, Ciurea SO. Haploidentical transplantation for acute myeloid leukemia patients with minimal/measurable residual disease at transplantation. Am J Hematol 2019; 94:1382-1387. [PMID: 31595538 DOI: 10.1002/ajh.25647] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/02/2019] [Accepted: 09/26/2019] [Indexed: 01/09/2023]
Abstract
There have been conflicting results regarding the impact of minimal/measurable disease at transplant on acute myeloid leukemia (AML) outcomes after haploidentical transplantation (haplo-SCT). We assessed the impact of pre-transplant disease status on post-transplant outcomes of 143 patients treated with haplo-SCT using fludarabine-melphalan (FM) conditioning and post-transplant cyclophosphamide (PTCy). With a median follow-up of 29 months, the two-year PFS for all patients was 41%. Compared to patients in complete remission (CR) at transplant, those with active disease (n = 29) and CR with incomplete count recovery (CRi) (n = 39) had worse PFS. They had hazard ratios (HR) of 3.5 (95% CI: 2.05-6.1; P < .001) and 2.3 (95% CI: 1.3-3.9; P = .002), respectively. Among patients who were in CR at transplant, there were no differences in PFS between those who had minimal residual disease (MRD) positive (n = 24), and MRD negative (n = 41) (HR 1.85, 95%CI: 0.9-4.0; P = .1). In multivariable analysis for patients in CR, only age was predictive for outcomes, while MRD status at transplant did not influence the treatment outcomes. Our findings suggest that haplo-SCT with FM conditioning regimen and PTCy-based GVHD prophylaxis has a protective effect, and may potentially abrogate the inferior outcomes of MRD positivity for patients with AML. Patients with positive MRD may benefit from proceeding urgently to a haplo-SCT, as this does not appear to negatively impact transplant outcomes.
Collapse
Affiliation(s)
- Samer A. Srour
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Rima M. Saliba
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Maria C. B. Bittencourt
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Jorge M. R. Perez
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Piyanuch Kongtim
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Gheath Al‐Atrash
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Amanda Olson
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Oran Betul
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Rohtesh Mehta
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Issa Khouri
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Lucia Masarova
- Department of Leukemia The University of Texas MD Anderson Cancer Center Houston Texas
| | - Nicholas Short
- Department of Leukemia The University of Texas MD Anderson Cancer Center Houston Texas
| | - Elias Jabbour
- Department of Leukemia The University of Texas MD Anderson Cancer Center Houston Texas
| | - Naval Daver
- Department of Leukemia The University of Texas MD Anderson Cancer Center Houston Texas
| | - Marina Konopleva
- Department of Leukemia The University of Texas MD Anderson Cancer Center Houston Texas
| | - Farhad Ravandi
- Department of Leukemia The University of Texas MD Anderson Cancer Center Houston Texas
| | - Hagop Kantarjian
- Department of Leukemia The University of Texas MD Anderson Cancer Center Houston Texas
| | - Richard E. Champlin
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| | - Stefan O. Ciurea
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston Texas
| |
Collapse
|
179
|
Bouvier A, Riou J, Thépot S, Sutra Del Galy A, François S, Schmidt A, Orvain C, Estienne MH, Villate A, Luque Paz D, Cottin L, Ribourtout B, Beucher A, Delneste Y, Ifrah N, Ugo V, Hunault-Berger M, Blanchet O. Quantitative chimerism in CD3-negative mononuclear cells predicts prognosis in acute myeloid leukemia patients after hematopoietic stem cell transplantation. Leukemia 2019; 34:1342-1353. [PMID: 31768015 DOI: 10.1038/s41375-019-0624-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/18/2019] [Accepted: 11/03/2019] [Indexed: 12/20/2022]
Abstract
Relapse is a major complication of acute myeloid leukemia (AML) after allogeneic hematopoietic stem cell transplantation (SCT). The objective of our study was to evaluate chimerism monitoring on the CD3-negative mononuclear cells by RQ-PCR to predict relapse of patients allografted for AML and to compare its performance with WT1 quantification. A cohort of 100 patients undergoing allogenic SCT for AML was retrospectively analyzed in a single institution. Patients without complete chimerism, defined as less than 0.01% of recipient's DNA in CD3-negative cells, had a significantly higher risk of relapse and a lower overall survival (p < 0.001). An increase in the percentage of recipient DNA in CD3-negative cells was associated with an increased risk of relapse (p < 0.001) but not with overall survival. Comparable performances between monitoring of CD3-negative cell chimerism and WT1 expression to predict relapse was observed up to more than 90 days before hematological relapse, with sensitivity of 82% and 78%, respectively, and specificity of 100% for both approaches. Quantitative specific chimerism of the CD3-negative mononuclear fraction, enriched in blastic cells, is a new and powerful tool for monitoring measurable residual disease and could be used for AML patients without available molecular markers.
Collapse
Affiliation(s)
- Anne Bouvier
- CHU Angers, Laboratoire d'Hématologie, Angers, France. .,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France. .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | - Jérémie Riou
- MINT, UMR INSERM 1066, CNRS 6021, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Sylvain Thépot
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | | | - Sylvie François
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Aline Schmidt
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Corentin Orvain
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Marie-Hélène Estienne
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Tours, Service d'Hématologie biologique, Tours, France
| | - Alban Villate
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Tours, Service d'Hématologie et thérapie cellulaire, Tours, France
| | - Damien Luque Paz
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Laurane Cottin
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Bénédicte Ribourtout
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France
| | - Annaëlle Beucher
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France
| | - Yves Delneste
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,CHU Angers, Laboratoire d'Immunologie et Allergologie, Angers, France
| | - Norbert Ifrah
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Valérie Ugo
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Mathilde Hunault-Berger
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Odile Blanchet
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Centre de Ressources Biologiques, BB-0033-00038, Angers, France
| |
Collapse
|
180
|
Nagler A, Dholaria B, Labopin M, Socie G, Huynh A, Itälä-Remes M, Deconinck E, Yakoub-Agha I, Cahn JY, Bourhis JH, Labussière-Wallet H, Chantepie S, Esteve J, Savani B, Mohty M. The impact of anti-thymocyte globulin on the outcomes of Patients with AML with or without measurable residual disease at the time of allogeneic hematopoietic cell transplantation. Leukemia 2019; 34:1144-1153. [PMID: 31728052 DOI: 10.1038/s41375-019-0631-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/14/2019] [Accepted: 11/03/2019] [Indexed: 02/03/2023]
Abstract
Measurable residual disease (MRD) status pre-allogeneic hematopoietic cell transplantation (allo-HCT) has been shown to predict transplant outcomes. We investigated the effect of Anti-Thymocyte Globulin (ATG) on acute myelogenous leukemia (AML) relapse by pretransplant MRD status. AML patients undergoing allo-HCT in first complete remission from either a matched sibling or unrelated donor during the 2006-2017 period were selected. Outcomes of 1509 patients (MRD+, n = 426) were studied. ATG was used in 561 (52%) and 239 (58%) patients within the MRD- and MRD+ cohorts, respectively. In MRD- patients, ATG did not affect relapse incidence (RI) (HR = 0.80, p = 0.17), but was associated with reduced incidence of grade II-IV acute GVHD, grade II-IV and chronic GVHD, reduced nonrelapse mortality (HR = 0.66, p = 0.05), improved leukemia-free survival (HR = 0.74, p = 0.02), overall survival (HR = 0.69, p = 0.01), and GVHD-relapse free survival (HR = 0.62, p < 0.01). In MRD+ patients, ATG was associated with a lower incidence of chronic GVHD (total, HR 0.56 p = 0.03; extensive, HR 0.40 P = 0.01), without an impact on other allo-HCT outcome parameters, including RI(HR = 1.02, p = 0.92). The use of ATG was associated with reduced risk for GVHD. ATG did not increase RI, even in high-risk AML patients who were MRD+ before allo-HCT.
Collapse
Affiliation(s)
- Arnon Nagler
- Hematology Department, Chaim Sheba Medical Center, Tel Hashomer, Israel.,Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, AP-HP, Sorobonne University, and INSERM UMRs 938, Paris, France
| | - Bhagirathbhai Dholaria
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Myriam Labopin
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, AP-HP, Sorobonne University, and INSERM UMRs 938, Paris, France
| | - Gerard Socie
- Bone Marrow Transplantation Department, CHU Saint-Louis, Paris, France
| | - Anne Huynh
- Oncopole, Institut Universitaire du Cancer Toulouse, Toulouse, France
| | - Maija Itälä-Remes
- Stem Cell Transplantation Unit, HUCH Comprehensive Cancer Center, Helsinki, Finland
| | - Eric Deconinck
- Service d'Hématologie, CHU Besançon, Besançon, France.,Inserm UMR 1098, Besançon, France
| | | | - Jean-Yves Cahn
- Service d'Hématologie, CHU Grenoble Alpes-Université Grenoble Alpes, Grenoble, France
| | - Jean-Henri Bourhis
- Division of Hematology, Department of Medical Oncology, Gustave Roussy, institut de cancérologie, BMT Service, Villejuif, France
| | - Hélène Labussière-Wallet
- Service Hématologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | | | - Jordi Esteve
- Hematology department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Bipin Savani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohamad Mohty
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, AP-HP, Sorobonne University, and INSERM UMRs 938, Paris, France
| |
Collapse
|
181
|
Jentzsch M, Schwind S, Bach E, Stasik S, Thiede C, Platzbecker U. Clinical Challenges and Consequences of Measurable Residual Disease in Non-APL Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:E1625. [PMID: 31652787 PMCID: PMC6893483 DOI: 10.3390/cancers11111625] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
The ability to detect residual levels of leukemic blasts (measurable residual disease, MRD) has already been integrated in the daily routine for treatment of patients with chronic myeloid and acute lymphoblastic leukemia. In acute myeloid leukemia (AML), a variety of mostly retrospective studies have shown that individuals in AML remission who tested positive for MRD at specific time-points or had increasing MRD levels are at significantly higher risk of relapse and death compared to MRD-negative patients. However, these studies differ with respect to the "MRD-target", time-point of MRD determination, material analyzed, and method applied. How this probably very valuable MRD information in individual patients may be adapted in the daily clinical routine, e.g., to separate patients who need more aggressive therapies from those who may be spared additional-potentially toxic-therapies is still a work-in-progress. With the exception of MRD assessment in acute promyelocytic leukemia (APL), the lack of randomized, prospective trials renders MRD-based decisions and clinical implications in AML a difficult task. As of today, we still do not have proof that early intervention in MRD-positive AML patients would improve outcomes, although this is very likely. In this article, we review the current knowledge on non-APL AML MRD assessment and possible clinical consequences.
Collapse
Affiliation(s)
- Madlen Jentzsch
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, Leipzig University Hospital, 04103 Leipzig, Germany.
| | - Sebastian Schwind
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, Leipzig University Hospital, 04103 Leipzig, Germany.
| | - Enrica Bach
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, Leipzig University Hospital, 04103 Leipzig, Germany.
| | - Sebastian Stasik
- Medical Department I, University Hospital and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany.
| | - Christian Thiede
- Medical Department I, University Hospital and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany.
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, Leipzig University Hospital, 04103 Leipzig, Germany.
| |
Collapse
|
182
|
Cho BS, Min GJ, Park SS, Shin SH, Yahng SA, Jeon YW, Yoon JH, Lee SE, Eom KS, Kim YJ, Lee S, Min CK, Cho SG, Kim DW, Lee JW, Kim M, Kim Y, Kim HJ. WT1 Measurable Residual Disease Assay in Patients With Acute Myeloid Leukemia Who Underwent Allogeneic Hematopoietic Stem Cell Transplantation: Optimal Time Points, Thresholds, and Candidates. Biol Blood Marrow Transplant 2019; 25:1925-1932. [DOI: 10.1016/j.bbmt.2019.05.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 11/18/2022]
|
183
|
PLCB4 upregulation is associated with unfavorable prognosis in pediatric acute myeloid leukemia. Oncol Lett 2019; 18:6057-6065. [PMID: 31788080 PMCID: PMC6865073 DOI: 10.3892/ol.2019.10921] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Phospholipase C (PLC) is a membrane-associated enzyme that regulates several cellular behaviors including cell motility, growth, transformation and differentiation. PLC is involved in cancer migration, invasion and drug resistance. However, the expression status and prognostic role of PLCB4 in acute myeloid leukemia (AML) remain unclear. In the present study, the complete clinical and mRNA expression data of 285 pediatric patients with de novo AML were obtained from the Therapeutically Available Research to Generate Effective Treatments database. The association between PLCB4 expression and clinical and molecular features was explored. The expression of PLCB4 was significantly higher in patients with AML who relapsed compared with those with long-term complete remission. Patients with PLCB4 upregulation had significantly lower overall survival (OS) and event free survival (EFS) rate compared with those with low PLCB4 expression. Multivariate Cox's regression analyses demonstrated that high PLCB4 expression was an independent risk factor of adverse OS (P<0.01; HR, 2.081) and EFS (P<0.01; HR, 2.130). Following stratification analysis according to transplant status in cases of first complete remission, the patients with high expression of PLCB4 had significantly lower OS and EFS rate in the chemotherapy group, but not the stem cell transplant group. Furthermore, PLCB4-associated genes were identified using Spearman's rank correlation analysis. KEGG pathway analysis revealed that PLCB4 and its associated genes were mainly involved in three potential pathways, including the Rap1 signaling pathway. Overall, the findings of the present study suggest that increased PLCB4 expression is associated with poor clinical outcome in pediatric patients with AML, and thus may represent a potential prognostic biomarker and therapeutic target for AML.
Collapse
|
184
|
Next-generation sequencing-based minimal residual disease monitoring in patients receiving allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia or myelodysplastic syndrome. Curr Opin Hematol 2019; 25:425-432. [PMID: 30281033 DOI: 10.1097/moh.0000000000000464] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW The monitoring of minimal residual disease (MRD) has important clinical implications in both the pre and postallogeneic stem cell transplant (SCT) setting in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Next-generation sequencing (NGS) is a rapidly improving technology whose application to the monitoring of MRD is an active area of research. We aim to describe existing methods of MRD in AML and MDS, with a focus on the utility of NGS in patients undergoing SCT. RECENT FINDINGS Flow cytometry and quantitative PCR have been recommended by the European Leukemia Net as the preferred methods of MRD in AML and MDS, but these methods have limitations in cases without a disease-defining phenotype and genotype. Clinical trials are currently ongoing to assess the use of NGS in the setting of SCT for MDS and AML. Few studies have so far assessed the optimal method of MRD monitoring in the posttransplant setting. SUMMARY The optimal method for the monitoring of MRD in AML and MDS both pre and post transplant may require more than one technology. NGS holds great promise for the monitoring of MRD, with prospective trials currently ongoing to evaluate its efficacy in this regard.
Collapse
|
185
|
Maurillo L, Bassan R, Cascavilla N, Ciceri F. Quality of Response in Acute Myeloid Leukemia: The Role of Minimal Residual Disease. Cancers (Basel) 2019; 11:cancers11101417. [PMID: 31548502 PMCID: PMC6826465 DOI: 10.3390/cancers11101417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022] Open
Abstract
In the acute myeloid leukemia (AML) setting, research has extensively investigated the existence and relevance of molecular biomarkers, in order to better tailor therapy with newly developed agents and hence improve outcomes and/or save the patient from poorly effective therapies. In particular, in patients with AML, residual disease after therapy does reflect the sum of the contributions of all factors associated with diagnosis and post-diagnosis resistance. The evaluation of minimal/measurable residual disease (MRD) can be considered as a key tool to guide patient’s management and a promising endpoint for clinical trials. In this narrative review, we discuss MRD evaluation as biomarker for tailored therapy in AML patients; we briefly report current evidence on the use of MRD in clinical practice, and comment on the potential ability of MRD in the assessment of the efficacy of new molecules.
Collapse
Affiliation(s)
- Luca Maurillo
- Hematology Unit, Department of Biomedicine and Prevention, Fondazione Policlinico Tor Vergata, Hospital, 00133 Rome, Italy.
| | - Renato Bassan
- Hematology Unit, dell'Angelo Hospital and Santissimi Giovanni and Paolo Hospital, 30174 Mestre and Venice, Italy.
| | - Nicola Cascavilla
- Hematology Unit, Onco-hematology Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy.
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, IRCCS S. Raffaele Scientific Institution, 20132 Milan, Italy.
| |
Collapse
|
186
|
Schuurhuis GJ, Ossenkoppele GJ, Kelder A, Cloos J. Measurable residual disease in acute myeloid leukemia using flow cytometry: approaches for harmonization/standardization. Expert Rev Hematol 2019; 11:921-935. [PMID: 30466339 DOI: 10.1080/17474086.2018.1549479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Measurable residual disease (MRD) in acute myeloid leukemia (AML) is a rapidly evolving area with many institutes embarking on it, both in academic and pharmaceutical settings. However, there is a multitude of approaches to design, perform, and report flow cytometric MRD. Together with the long-term experience needed, this makes flow cytometric MRD in AML nonstandardized and time-consuming. Areas covered: This paper briefly summarizes critical issues, like sample preparation and transport, markers and fluorochromes of choice, but in particular focuses on the main issues, which includes specificity and sensitivity, hereby providing a new model that may circumvent the main disadvantages of the present approaches. New approaches that may add to the value of flow cytometric MRD includes assessment of leukemia stem cells, MRD in peripheral blood, and approaches to use multidimensional image analysis. Expert commentary: MRD in AML requires standardization/harmonization on many aspects, for which the present paper offers possible guidelines.
Collapse
Affiliation(s)
- Gerrit J Schuurhuis
- a Department of Hematology , VU University Medical Center , Amsterdam , Netherlands
| | - Gert J Ossenkoppele
- a Department of Hematology , VU University Medical Center , Amsterdam , Netherlands
| | - Angèle Kelder
- a Department of Hematology , VU University Medical Center , Amsterdam , Netherlands
| | - Jacqueline Cloos
- a Department of Hematology , VU University Medical Center , Amsterdam , Netherlands
| |
Collapse
|
187
|
GIMEMA AML1310 trial of risk-adapted, MRD-directed therapy for young adults with newly diagnosed acute myeloid leukemia. Blood 2019; 134:935-945. [DOI: 10.1182/blood.2018886960] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
Key Points
A risk-adapted, MRD-driven transplant strategy is a feasible approach for the treatment of younger adults with AML. Pretransplant MRD positivity should not contraindicate delivery of an allogeneic stem cell transplant.
Collapse
|
188
|
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: 6.0] [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
|
189
|
Selim A, Alvaro F, Cole CH, Fraser CJ, Mechinaud F, O'Brien TA, Shaw PJ, Tapp H, Teague L, Nivison-Smith I, Moore AS. Hematopoietic stem cell transplantation for children with acute myeloid leukemia in second remission: A report from the Australasian Bone Marrow Transplant Recipient Registry and the Australian and New Zealand Children's Haematology Oncology Group. Pediatr Blood Cancer 2019; 66:e27812. [PMID: 31111633 DOI: 10.1002/pbc.27812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/10/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Approximately one-third of children with acute myeloid leukemia (AML) relapse, requiring re-treatment and allogeneic hematopoietic stem cell transplantation (HSCT). Although achieving second complete remission (CR2) prior to HSCT is desirable, once CR2 is attained, it is unclear if there is any benefit from further chemotherapy prior to HSCT. Moreover, although pre-HSCT minimal residual disease (MRD) has prognostic value in acute lymphoblastic leukemia, the benefit of MRD reduction after achieving CR prior to HSCT is less clear for AML. PROCEDURE To address these questions, we analyzed data from pediatric transplant centers in Australia and New Zealand concerning relapsed childhood AML cases occurring between 1998 and 2013. Given the retrospective nature of our analysis and assay data available, we analyzed patients on the basis of measurable residual disease (MeRD) by any methodology, rather than MRD in the conventional sense. RESULTS We observed improved overall survival (OS) in children receiving two chemotherapy cycles, compared to one cycle or three or more cycles pre-HSCT. Improved OS with two cycles remained significant for patients without MeRD after cycle 1. CONCLUSIONS These data suggest that a second chemotherapy cycle pre-HSCT may improve survival by lowering disease burden. Prospective trials assessing strategies to reduce pre-HSCT MRD in relapsed childhood AML are warranted.
Collapse
Affiliation(s)
- Adrian Selim
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | | | - Catherine H Cole
- Department of Haematology/Oncology, Princess Margaret Hospital for Children, Perth, Australia
| | - Chris J Fraser
- Oncology Services Group, Queensland Children's Hospital, Brisbane, Australia
| | | | - Tracey A O'Brien
- Kids' Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Peter J Shaw
- Oncology Unit, The Children's Hospital at Westmead, Sydney, Australia
| | - Heather Tapp
- Department of Clinical Haematology/Oncology, Women's and Children's Hospital, Adelaide, Australia
| | | | - Ian Nivison-Smith
- Australasian Bone Marrow Transplant Recipient Registry, Sydney, Australia
| | - Andrew S Moore
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Oncology Services Group, Queensland Children's Hospital, Brisbane, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
190
|
Chronic graft-versus-host disease could ameliorate the impact of adverse somatic mutations in patients with myelodysplastic syndromes and hematopoietic stem cell transplantation. Ann Hematol 2019; 98:2151-2162. [DOI: 10.1007/s00277-019-03751-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 06/27/2019] [Indexed: 11/28/2022]
|
191
|
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: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/08/2019] [Indexed: 12/29/2022]
|
192
|
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: 0.8] [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
|
193
|
Abstract
PURPOSE OF REVIEW Incorporation of minimal residual disease (MRD) testing in acute lymphoblastic leukemia (ALL) and acute myeloblastic leukemia (AML) has transformed the landscape of hematopoietic cell transplantation (HCT). Pre-HCT MRD has allowed prognostication of HCT outcomes for high-risk leukemia patients, whereas the detection of post-HCT MRD has allowed for interventions to decrease relapse. RECENT FINDINGS In this review, we emphasize studies from the past two decades that highlight the critical role of MRD in HCT in pediatric ALL and AML. Advances in MRD detection methodology, using next-generation sequencing, have improved the sensitivity of MRD testing allowing for more accurate predictions of HCT outcomes for patients with relapsed and refractory ALL and AML. In addition, novel pre-HCT therapies, especially immunotherapy in ALL, have dramatically increased the number of patients who achieve MRD-negative remissions pre-HCT, resulting in improved HCT outcomes. Post-HCT MRD remains a challenge and new therapeutic interventions are needed to reduce post-HCT relapse. SUMMARY As immunotherapy increases pre-HCT MRD-negative remissions, and next-generation sequencing-MRD is incorporated to improve the sensitivity of MRD detection, future clinical studies will investigate less toxic HCT approaches to reduce long-term sequelae and to identify which patients may benefit most from early post-HCT intervention to reduce relapse.
Collapse
Affiliation(s)
- Agne Taraseviciute
- Division of Pediatric Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | | |
Collapse
|
194
|
Wong HY, Sung AD, Lindblad KE, Sheela S, Roloff GW, Rizzieri D, Goswami M, Mulé MP, Ramos NR, Tang J, Thompson J, DeStefano CB, Romero K, Dillon LW, Kim DY, Lai C, Hourigan CS. Molecular Measurable Residual Disease Testing of Blood During AML Cytotoxic Therapy for Early Prediction of Clinical Response. Front Oncol 2019; 8:669. [PMID: 30697529 PMCID: PMC6341003 DOI: 10.3389/fonc.2018.00669] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
Measurable residual disease (MRD) testing after initial chemotherapy treatment can predict relapse and survival in acute myeloid leukemia (AML). However, it has not been established if repeat molecular or genetic testing during chemotherapy can offer information regarding the chemotherapy sensitivity of the leukemic clone. Blood from 45 adult AML patients at day 1 and 4 of induction (n = 35) or salvage (n = 10) cytotoxic chemotherapy was collected for both quantitative real-time PCR (qPCR) assessment (WT1) and next generation sequencing (>500 × depth) of 49 gene regions recurrently mutated in MDS/AML. The median age of subjects was 62 (23–78); 42% achieved a complete response. WT1 was overexpressed in most patients tested but was uninformative for very early MRD assessment. A median of 4 non-synonymous variants (range 0–7) were detected by DNA sequencing of blood on day 1 of therapy [median variant allele frequency (VAF): 29%]. Only two patients had no variants detectable. All mutations remained detectable in blood on day 4 of intensive chemotherapy and remarkably the ratio of mutated to wild-type sequence was often maintained. This phenomenon was not limited to variants in DNMT3A, TET2, and ASXL1. The kinetics of NPM1 and TP53 variant burden early during chemotherapy appeared to be exceptions and exhibited consistent trends in this cohort. In summary, molecular testing of blood on day 4 of chemotherapy is not predictive of clinical response to cytotoxic induction therapy in AML. The observed stability in variant allele frequency suggests that cytotoxic therapy may have a limited therapeutic index for clones circulating in blood containing these mutations. Further validation is required to confirm the utility of monitoring NPM1 and TP53 kinetics in blood during cytotoxic therapy.
Collapse
Affiliation(s)
- Hong Yuen Wong
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Anthony D Sung
- Duke University School of Medicine, Durham, NC, United States
| | - Katherine E Lindblad
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Sheenu Sheela
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Gregory W Roloff
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - David Rizzieri
- Duke University School of Medicine, Durham, NC, United States
| | - Meghali Goswami
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Matthew P Mulé
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Nestor R Ramos
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Jingrong Tang
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Julie Thompson
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Christin B DeStefano
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Kristi Romero
- Duke University School of Medicine, Durham, NC, United States
| | - Laura W Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Dong-Yun Kim
- Office of Biostatistics Research, Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Catherine Lai
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| |
Collapse
|
195
|
Abu-Arja RF, Dargart JL, Bajwa RPS, Kahwash SB, Auletta JJ, Rangarajan HG. Monozygotic twins diagnosed simultaneously with RAM immunophenotype acute myeloid leukemia. Pediatr Transplant 2018; 22:e13291. [PMID: 30220110 DOI: 10.1111/petr.13291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 12/31/2022]
Abstract
AML with the RAM immunophenotype is associated with extremely poor prognosis. We report a rare case of monozygotic twins presenting simultaneously at the age of 2 years with RAM AML. Each twin underwent a myeloablative 7/10 unrelated umbilical cord blood transplant. Pretransplant Twin A's bone marrow was negative for MRD by flow cytometry (<0.01%) unlike Twin B's bone marrow (0.07%). Twin A is alive in remission 3 years from transplant. Twin B developed primary graft failure, but subsequently rescued with a haploidentical stem cell transplant. However, she relapsed and died 13 months from diagnosis. The twins' clinical courses demonstrate that upfront intensive chemotherapy to achieve negative MRD, followed by allogeneic hematopoietic stem cell transplant as postremission intensification strategy, should be considered in this high-risk AML.
Collapse
Affiliation(s)
- Rolla F Abu-Arja
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Jamie L Dargart
- Department of Pediatric Hematology and Oncology, ProMedica Toledo Children's Hospital, Toledo, Ohio
| | - Rajinder P S Bajwa
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Samir B Kahwash
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Jeffery J Auletta
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Hemalatha G Rangarajan
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
196
|
Pollyea DA. New drugs for acute myeloid leukemia inspired by genomics and when to use them. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:45-50. [PMID: 30504290 PMCID: PMC6245963 DOI: 10.1182/asheducation-2018.1.45] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We are several years into the "postdiscovery" era in acute myeloid leukemia (AML) thanks to extensive work involving the sequencing of genomes and exomes of countless patients, which has led to routine comprehensive targeted sequencing in clinical care. The ability to unlock the molecular underpinnings of each patient's disease was supposed to usher in a new treatment era in which each patient was assigned, based on her mutational profile, a personalized cocktail of targeted therapies that would snuff the disease into submission with minimal toxicity. Whether we have fully realized the promise of personalized therapy in AML is unclear. Here, I review those new drugs that have been inspired by genomics, discuss others that might be possible and their potential roles, and consider whether the ability to target genomic mutations in a personalized manner constitutes the future of AML therapeutics or is representative of an era that has already passed.
Collapse
Affiliation(s)
- Daniel A Pollyea
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
197
|
Lee YJ, Baek DW, Ahn JS, Ahn SY, Jung SH, Yang DH, Lee JJ, Kim HJ, Ham JY, Suh JS, Sohn SK, Moon JH. Impact of Consolidation Cycles Before Allogeneic Hematopoietic Cell Transplantation for Acute Myeloid Leukemia in First Complete Remission. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:e529-e535. [PMID: 30268640 DOI: 10.1016/j.clml.2018.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/12/2018] [Accepted: 08/21/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND The optimal number of high-dose cytarabine (HDAC) consolidation cycles before allogeneic hematopoietic cell transplantation (HCT) for acute myeloid leukemia is not fully standardized. PATIENTS AND METHODS This study evaluated the impact of HDAC consolidation cycles before allogeneic HCT in 194 patients with acute myeloid leukemia in first complete remission between 1998 and 2014. The patients were reclassified into 3 groups-no consolidation (C0, n = 20), 1 consolidation (C1, n = 115), and ≥ 2 consolidations (C2, n = 59)-by pre-HCT consolidation cycle. RESULTS The 3-year relapse-free survival rates was 45.9%, 66.9%, and 73.3% for the C0, C1, and C2 groups, respectively (P = .064), while the 3-year overall survival rates were 35.0%, 55.2%, and 67.5%, respectively (P = .106). The cumulative incidence of acute graft-versus-host disease (GVHD) was higher in the C2 group (38.7%) than in the C0 (22.2%) or C1 (17.7%) group (P = .018). However, the incidence of chronic GVHD showed no difference between the groups. Multivariate analysis for overall survival revealed the following independent factors: adverse cytogenetic risk (hazard ratio [HR] = 1.84, P = .046), C2 versus C0 (HR = 0.41, P = .037), pre-HCT status beyond CR1 versus CR1 (HR = 5.78, P < .001), and presence of chronic GVHD (HR = 0.45, P = .004). CONCLUSION One or two cycles of HDAC consolidation therapy led to better subsequent HCT outcomes compared to the no-consolidation therapy group.
Collapse
Affiliation(s)
- Yoo Jin Lee
- Department of Hematology/Oncology, Kyungpook National University Hospital, Daegu, Korea; School of Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Dong Won Baek
- Department of Hematology/Oncology, Kyungpook National University Hospital, Daegu, Korea; School of Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Jae-Sook Ahn
- Department of Hematology/Oncology, Chonnam National University Hwasun Hospital, Jeollanam-do, Korea
| | - Seo-Yeon Ahn
- Department of Hematology/Oncology, Chonnam National University Hwasun Hospital, Jeollanam-do, Korea
| | - Sung-Hoon Jung
- Department of Hematology/Oncology, Chonnam National University Hwasun Hospital, Jeollanam-do, Korea
| | - Deok-Hwan Yang
- Department of Hematology/Oncology, Chonnam National University Hwasun Hospital, Jeollanam-do, Korea
| | - Je-Jung Lee
- Department of Hematology/Oncology, Chonnam National University Hwasun Hospital, Jeollanam-do, Korea
| | - Hyeoung Joon Kim
- Department of Hematology/Oncology, Chonnam National University Hwasun Hospital, Jeollanam-do, Korea
| | - Ji Yeon Ham
- School of Medicine, Kyungpook National University Hospital, Daegu, Korea; Department of Laboratory Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Jang Soo Suh
- School of Medicine, Kyungpook National University Hospital, Daegu, Korea; Department of Laboratory Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Sang Kyun Sohn
- Department of Hematology/Oncology, Kyungpook National University Hospital, Daegu, Korea; School of Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Joon Ho Moon
- Department of Hematology/Oncology, Kyungpook National University Hospital, Daegu, Korea; School of Medicine, Kyungpook National University Hospital, Daegu, Korea.
| |
Collapse
|
198
|
Burnett AK, Das Gupta E, Knapper S, Khwaja A, Sweeney M, Kjeldsen L, Hawkins T, Betteridge SE, Cahalin P, Clark RE, Hills RK, Russell NH. Addition of the mammalian target of rapamycin inhibitor, everolimus, to consolidation therapy in acute myeloid leukemia: experience from the UK NCRI AML17 trial. Haematologica 2018; 103:1654-1661. [PMID: 29976746 PMCID: PMC6165825 DOI: 10.3324/haematol.2018.189514] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/04/2018] [Indexed: 01/14/2023] Open
Abstract
As part of the UK NCRI AML17 trial, adult patients with acute myeloid leukemia in remission could be randomized to receive the mammalian target of rapamycin inhibitor everolimus, sequentially with post-induction chemotherapy. Three hundred and thirty-nine patients were randomised (2:1) to receive everolimus or not for a maximum of 84 days between chemotherapy courses. The primary endpoint was relapse-free survival. At 5 years there was no difference in relapse-free survival [29% versus 40%; odds ratio 1.19 (0.9-1.59) P=0.2], cumulative incidence of relapse [60% versus 54%: odds ratio 1.12 (0.82-1.52): P=0.5] or overall survival [45% versus 58%: odds ratio 1.3 (0.94-1.81): P=0.11]. The independent Data Monitoring Committee advised study termination after randomization of 339 of the intended 600 patients because of excess mortality in the everolimus arm without any evidence of beneficial disease control. The delivery of the everolimus dose was variable, but there was no evidence of clinical benefit in patients with adequate dose delivery compared with no treatment. This study suggests that the addition of mammalian target of rapamycin inhibition to chemotherapy provides no benefit.
Collapse
Affiliation(s)
- Alan K Burnett
- Formerly Department of Haematology, Cardiff University School of Medicine, UK
| | - Emma Das Gupta
- Department of Haematology, Nottingham University Hospital NHS Trust, UK
| | - Steve Knapper
- Department of Haematology, University Hospital of Wales, Cardiff, UK
| | - Asim Khwaja
- University College, London Cancer Institute, UK
| | - Marion Sweeney
- Department of Haematology, University Hospital of Wales, Cardiff, UK
| | - Lars Kjeldsen
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark
| | - Timothy Hawkins
- Department of Haematology, Auckland City Hospital, New Zealand
| | | | - Paul Cahalin
- Department of Haematology, Blackpool Victoria Hospital, UK
| | - Richard E Clark
- Department of Haematology, Royal Liverpool University Hospital, UK
| | - Robert K Hills
- Department of Haematology, Royal Liverpool University Hospital, UK
| | - Nigel H Russell
- Department of Haematology, Nottingham University Hospital NHS Trust, UK
| |
Collapse
|
199
|
Buccisano F, Maurillo L, Schuurhuis GJ, Del Principe MI, Di Veroli A, Gurnari C, Venditti A. The emerging role of measurable residual disease detection in AML in morphologic remission. Semin Hematol 2018; 56:125-130. [PMID: 30926088 DOI: 10.1053/j.seminhematol.2018.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/03/2018] [Accepted: 09/14/2018] [Indexed: 01/08/2023]
Abstract
Despite the increasing knowledge of the genomic landscape of acute myeloid leukemia (AML), prediction merely based on genetics fails to anticipate outcome, presumably due to the heterogeneous composition of the leukemic clone determining complex interactions between different genetic abnormalities. Therefore, the introduction of a post-treatment biomarker exploring the quality of response to therapy such as assessment of measurable (previously minimal) residual disease (MRD) may lead to refinements of the prognostic assessment in AML. In this view, the European LeukemiaNet has recently endorsed the achievement of a MRD negative morphologic complete remission as a purpose the treatment. Techniques like multiparametric flow cytometry and reverse transcriptase-quantitative polymerase chain reaction have reached a level of sensitivity and specificity that make them ready for introduction in clinical practice. In the present review, we will give an update on the efforts in harmonization and/or standardization of MRD assessment in AML, focusing on the newest acquisitions in the clinical applications of MRD, and considering issues like relationship of MRD with leukemic stem cells or MRD assessment in peripheral blood.
Collapse
Affiliation(s)
- F Buccisano
- Hematology Unit, Department of Biomedicine and Prevention, University tor Vergata of Rome, Rome, Italy.
| | - L Maurillo
- Hematology Unit, Department of Biomedicine and Prevention, University tor Vergata of Rome, Rome, Italy
| | - G J Schuurhuis
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - M I Del Principe
- Hematology Unit, Department of Biomedicine and Prevention, University tor Vergata of Rome, Rome, Italy
| | - A Di Veroli
- Hematology Unit, Department of Biomedicine and Prevention, University tor Vergata of Rome, Rome, Italy
| | - C Gurnari
- Hematology Unit, Department of Biomedicine and Prevention, University tor Vergata of Rome, Rome, Italy
| | - A Venditti
- Hematology Unit, Department of Biomedicine and Prevention, University tor Vergata of Rome, Rome, Italy
| |
Collapse
|
200
|
Gilleece MH, Labopin M, Yakoub-Agha I, Volin L, Socié G, Ljungman P, Huynh A, Deconinck E, Wu D, Bourhis JH, Cahn JY, Polge E, Mohty M, Savani BN, Nagler A. Measurable residual disease, conditioning regimen intensity, and age predict outcome of allogeneic hematopoietic cell transplantation for acute myeloid leukemia in first remission: A registry analysis of 2292 patients by the Acute Leukemia Working Party European Society of Blood and Marrow Transplantation. Am J Hematol 2018; 93:1142-1152. [PMID: 29981272 DOI: 10.1002/ajh.25211] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 12/26/2022]
Abstract
Patients with acute myeloid leukemia (AML) in morphological first complete remission (CR1) pre-allogeneic hematopoietic cell transplantation (HCT) may have measurable residual disease (MRD) by molecular and immunophenotyping criteria. We assessed interactions of MRD status with HCT conditioning regimen intensity in patients aged <50 years (y) or ≥50y. This was a retrospective study by the European Society for Blood and Marrow Transplantation registry. Patients were >18y with AML CR1 MRD NEG/POS and recipients of HCT in 2000-2015. Conditioning regimens were myeloablative (MAC), reduced intensity (RIC) or non-myeloablative (NMA). Outcomes included leukemia free survival (LFS), overall survival (OS), relapse incidence (RI), non-relapse mortality (NRM), chronic graft-vs-host (cGVHD), and GVHD-free and relapse-free survival (GRFS). The 2292 eligible patients were categorized into four paired groups: <50y MRD POS MAC (N = 240) vs RIC/NMA (N = 58); <50y MRD NEG MAC (N = 665) vs RIC/NMA (N = 195); ≥50y MRD POS MAC (N = 126) vs RIC/NMA (N = 230), and ≥50y MRD NEG MAC (N = 223) vs RIC/NMA (N = 555). In multivariate analysis RIC/NMA was only inferior to MAC for patients in the <50y MRD POS group, with worse RI (HR 1.71) and LFS (HR 1.554). Patients <50Y MRD NEG had less cGVHD after RIC/NMA HCT (HR 0.714). GRFS was not significantly affected by conditioning intensity in any group. Patients aged <50y with AML CR1 MRD POS status should preferentially be offered MAC allo-HCT. Prospective studies are needed to address whether patients with AML CR1 MRD NEG may be spared the toxicity of MAC regimens. New approaches are needed for ≥50y AML CR1 MRD POS.
Collapse
Affiliation(s)
- Maria H. Gilleece
- Department of Haematology; Leeds Teaching Hospitals Trust, University of Leeds; Leeds United Kingdom
| | | | | | - Liisa Volin
- Comprehensive Cancer Center, Stem Cell Transplantation Unit; Helsinki University Hospital; Helsinki Finland
| | - Gerard Socié
- Service d'Hématologie Greffe; Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris; Paris France
| | - Per Ljungman
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital; Stockholm Sweden
| | - Anne Huynh
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse Oncopole; Toulouse France
| | - Eric Deconinck
- Hematology Department; CHRU Besancon, INSERM UMR1098, Universite de Franche-Comte; Besancon France
| | - Depei Wu
- Department of Hematology; First Affiliated Hospital of Soochow University; Suzhou Jiangsu China
| | | | - Jean Yves Cahn
- Department of Haematology, Centre Hospital; Universitaire Grenoble Alpes; Grenoble France
| | - Emmanuelle Polge
- Acute Leukemia Working Party; European Society for Blood and Marrow Transplantation Paris Study Office/European Center for Biostatistical and Epidemiological Evaluation in Hematopoietic Cell Therapy (CEREST-TC); Paris France
| | - Mohamad Mohty
- Hopital Saint-Antoine, Université Pierre and Marie Curie, Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche U938; Paris France
| | - Bipin N. Savani
- Division of Hematology/Oncology, Department of Internal Medicine; Vanderbilt University Medical Center; Nashville Tennessee
| | - Arnon Nagler
- Chaim Sheba Medical Center; Tel Aviv University; Tel-Hashomer Israel
| |
Collapse
|