101
|
Khanolkar RA, Faridi RM, Kinzel M, Jamani K, Savoie ML, Shafey M, Khan FM, Storek J. Impact of FLT3 internal tandem duplication and NPM1 mutations in acute myeloid leukemia treated with allogeneic hematopoietic cell transplantation. Cytotherapy 2021; 24:413-420. [PMID: 34863627 DOI: 10.1016/j.jcyt.2021.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/06/2021] [Accepted: 10/06/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AIMS The internal tandem duplication of FLT3 (FLT3ITD) and NPM1 mutations (NPM1mut) are well-established prognostic factors in cytogenetically intermediate-risk acute myeloid leukemia (AML) when treated with chemotherapy alone. However, their prognostic value in the setting of allogeneic hematopoietic cell transplantation (HCT) is controversial. METHODS FLT3 and NPM1 mutational status was determined at diagnosis using single-gene polymerase chain reaction or next-generation sequencing in 247 adult patients with cytogenetically intermediate-risk AML who underwent myeloablative HCT. Multivariate Fine-Gray and Cox regression was used to analyze the cumulative incidence of relapse (CIR), relapse-free survival (RFS) and overall survival (OS). RESULTS FLT3ITD and NPM1mut were present in 74 of 247 (30%) and 79 of 247 (32%) patients, respectively. There was no significant difference between patients without a FLT3ITD or NPM1mut (FLT3NONITD/NPM1WT) and patients with a FLT3ITD mutation alone (FLT3ITD/NPM1WT) with regard to CIR (P = 0.60), RFS (P = 0.91) or OS (P = 0.66). Similarly, there was no significant difference between FLT3NONITD/NPM1WT and FLT3NONITD/NPM1mut patients with regard to CIR (P = 0.70), RFS (P = 0.75) or OS (P = 0.95). The presence of a concurrent mutation in NPM1 did not appear to modify the impact of having a FLT3ITD mutation. CONCLUSIONS In contrast to chemotherapy-only treatment, FLT3 and NPM1 mutational status does not appear to predict outcomes in patients with cytogenetically intermediate-risk AML following HCT. These results suggest that HCT may ameliorate the poor prognostic effect of FLT3ITD mutation and that HCT should be considered over chemotherapy-only treatment in FLT3ITD-mutated AML.
Collapse
Affiliation(s)
| | - Rehan M Faridi
- Cumming School of Medicine, University of Calgary, Calgary, Canada; Alberta Precision Laboratories, Calgary, Canada
| | - Megan Kinzel
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Kareem Jamani
- Cumming School of Medicine, University of Calgary, Calgary, Canada; Alberta Health Services, Calgary, Canada
| | - Mary L Savoie
- Cumming School of Medicine, University of Calgary, Calgary, Canada; Alberta Health Services, Calgary, Canada
| | - Mona Shafey
- Cumming School of Medicine, University of Calgary, Calgary, Canada; Alberta Health Services, Calgary, Canada
| | - Faisal M Khan
- Cumming School of Medicine, University of Calgary, Calgary, Canada; Alberta Precision Laboratories, Calgary, Canada
| | - Jan Storek
- Cumming School of Medicine, University of Calgary, Calgary, Canada; Alberta Health Services, Calgary, Canada
| |
Collapse
|
102
|
Jimenez Jimenez AM, De Lima M, Komanduri KV, Wang TP, Zhang MJ, Chen K, Abdel-Azim H, Abid MB, Aljurf M, Alkhateeb H, Assal A, Bacher U, Baron F, Battiwalla M, Beitinjaneh A, Bejanyan N, Bhatt VR, Byrne M, Cahn JY, Cairo M, Castillo P, Copelan E, DeFilipp Z, Perez MAD, Elsawy M, Gale RP, George B, Grunwald MR, Hildebrandt GC, Hogan WJ, Kanakry CG, Kansagra A, Kharfan-Dabaja MA, Khera N, Krem MM, Lazaryan A, Maakaron J, Martino R, McGuirk J, Michelis FV, Milone G, Mishra A, Murthy HS, Mussetti A, Nathan S, Nishihori T, Olsson RF, Palmisiano N, Patel S, Saad A, Seo S, Sharma A, Solh M, Verdonck LF, Wirk B, Yared JA, Litzow M, Kebriaei P, Hourigan CS, Saber W, Weisdorf D. An adapted European LeukemiaNet genetic risk stratification for acute myeloid leukemia patients undergoing allogeneic hematopoietic cell transplant. A CIBMTR analysis. Bone Marrow Transplant 2021; 56:3068-3077. [PMID: 34584240 PMCID: PMC8758206 DOI: 10.1038/s41409-021-01450-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/31/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022]
Abstract
Cytogenetic and molecular abnormalities are known to influence post-transplant outcomes in acute myeloid leukemia (AML) but data assessing the prognostic value of combined genetic models in the HCT setting are limited. We developed an adapted European LeukemiaNet (aELN) risk classification based on available genetic data reported to the Center for International Blood and Marrow Transplant Research, to predict post-transplant outcomes in 2289 adult AML patients transplanted in first remission, between 2013 and 2017. Patients were stratified according to aELN into three groups: favorable (Fav, N = 181), intermediate (IM, N = 1185), and adverse (Adv, N = 923). Univariate analysis demonstrated significant differences in 2-year overall survival (OS) (Fav: 67.7%, IM: 64.9% and Adv: 53.9%; p < 0.001); disease-free survival (DFS) (Fav: 57.8%, IM: 55.5% and Adv: 45.3; p < 0.001) and relapse (Fav: 28%, IM: 27.5% and Adv: 37.5%; p < 0.001). Multivariate analysis (MVA) revealed no differences in outcomes between the Fav and IM groups, thus they were combined. On MVA, patients in the Adv risk group had the highest risk of relapse (HR 1.47 p ≤ 0.001) and inferior DFS (HR 1.35 p < 0.001) and OS (HR 1.39 p < 0.001), even using myeloablative conditioning or in those without the pre-HCT measurable-residual disease. Novel approaches to mitigate relapse in this high-risk group are urgently needed.
Collapse
Affiliation(s)
- Antonio M Jimenez Jimenez
- Division of Transplantation and Cellular Therapy, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Marcos De Lima
- Department of Medicine, Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Krishna V Komanduri
- Division of Transplantation and Cellular Therapy, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Trent P Wang
- Division of Transplantation and Cellular Therapy, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mei-Jie Zhang
- (CIBMTR)® Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Karen Chen
- (CIBMTR)® Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hisham Abdel-Azim
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Muhammad Bilal Abid
- Divisions of Hematology/Oncology & Infectious Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mahmoud Aljurf
- Department of Oncology, King Faisal Specialist Hospital Center & Research, Riyadh, Saudi Arabia
| | | | - Amer Assal
- Columbia University Irving Medical Center, Department of Medicine, Bone Marrow Transplant and Cell Therapy Program, New York, NY, USA
| | - Ulrike Bacher
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | | | - Amer Beitinjaneh
- Division of Transplantation and Cellular Therapy, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nelli Bejanyan
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, FL, USA
| | - Vijaya Raj Bhatt
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael Byrne
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jean-Yves Cahn
- Department of Hematology, CHU Grenoble Alpes, Université Grenoble Alpes, Grenoble, France
| | - Mitchell Cairo
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Paul Castillo
- UF Health Shands Children's Hospital, Gainesville, FL, USA
| | - Edward Copelan
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Zachariah DeFilipp
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, MA, USA
| | - Miguel Angel Diaz Perez
- Department of Hematology/Oncology, Hospital Infantil Universitario Niño Jesus, Madrid, Spain
| | - Mahmoud Elsawy
- Division of Hematology, Department of Medicine, Dalhousie University, Halifax, Canada
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunology and Inflammation, Imperial College London, London, UK
| | | | - Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | | | | | - Christopher G Kanakry
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ankit Kansagra
- UT Southwestern Medical Center, Blood and Marrow Transplant Program, Dallas, TX, USA
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Nandita Khera
- Department of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Maxwell M Krem
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Aleksandr Lazaryan
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, FL, USA
| | - Joseph Maakaron
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rodrigo Martino
- Divison of Clinical Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Joseph McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Minneapolis, MN, USA
| | - Fotios V Michelis
- Allogeneic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Giuseppe Milone
- Azienda Ospedaliera Universitaria Policlinico-San Marco, Catania, Italy
| | - Asmita Mishra
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, FL, USA
| | - Hemant S Murthy
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Alberto Mussetti
- Hematology Department, Institut Catalá d' Oncologia-Hospitalet, Barcelona, Spain
- IDIBELL-Institut Catalá d' Oncologia, l'Hospitalet de Llobregat, El Prat de Llobregat, Spain
| | - Sunita Nathan
- Section of Bone Marrow Transplant and Cell Therapy, Rush University Medical Center, Chicago, IL, USA
| | - Taiga Nishihori
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, FL, USA
| | - Richard F Olsson
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Clinical Research Sormland, Uppsala University, Uppsala, Sweden
| | - Neil Palmisiano
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sagar Patel
- Blood and Marrow Transplant Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Ayman Saad
- Division of Hematology, Ohio State University, Columbus, OH, USA
| | - Sachiko Seo
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, GA, USA
| | - Leo F Verdonck
- Department of Hematology/Oncology, Isala Clinic, Zwolle, the Netherlands
| | - Baldeep Wirk
- Bone Marrow Transplant Program, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Jean A Yared
- Blood & Marrow Transplantation Program, Division of Hematology/Oncology, Department of Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA
| | - Mark Litzow
- Division of Hematology and Transplant Center, Mayo Clinic Rochester, Rochester, MN, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute National Institutes of Health, Bethesda, MD, USA
| | - Wael Saber
- (CIBMTR)® Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Daniel Weisdorf
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
103
|
Clofarabine-fludarabine-busulfan in HCT for pediatric leukemia: an effective, low toxicity, TBI-free conditioning regimen. Blood Adv 2021; 6:1719-1730. [PMID: 34781362 PMCID: PMC8941455 DOI: 10.1182/bloodadvances.2021005224] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 11/20/2022] Open
Abstract
CloFluBu-conditioning results in encouraging EFS for ALL and AML, with low TRM, limited incidence of aGvHD and GF, and no cases of VOD. Minimal residual disease status prior to transplantation impacted outcome due to increased relapse risk in both AML and ALL patients.
We prospectively studied clofarabine-fludarabine-busulfan (CloFluBu)-conditioning in allogeneic hematopoietic cell therapy (HCT) for lymphoid and myeloid malignancies and hypothesized that CloFluBu provides a less toxic alternative to conventional conditioning regimens, with adequate antileukemic activity. All patients receiving their first HCT, from 2011-2019, were included and received CloFluBu. The primary endpoint was event-free survival (EFS). Secondary endpoints were overall survival (OS), graft-versus-host disease (GvHD)-relapse-free survival (GRFS), treatment-related mortality (TRM), cumulative incidence of relapse (CIR), acute and chronic GvHD (aGvHD and cGvHD), and veno-occlusive disease (VOD). Cox proportional hazard and Fine and Gray competing-risk models were used for data analysis. One hundred fifty-five children were included: 60 acute lymphoid leukemia (ALL), 69 acute myeloid leukemia (AML), and 26 other malignancies (mostly MDS-EB). The median age was 9.7 (0.5 to 18.6) years. Estimated 2-year EFS was 72.0% ± 6.0 in ALL patients, and 62.4% ± 6.0 in AML patients. TRM in the whole cohort was 11.0% ± 2.6, incidence of aGvHD 3 to 4 at 6 months was 12.3% ± 2.7, extensive cGvHD at 2 years was 6.4% ± 2.1. Minimal residual disease-positivity prior to HCT was associated with higher CIR, both in ALL and AML. CloFluBu showed limited toxicity and encouraging EFS. CloFluBu is a potentially less toxic alternative to conventional conditioning regimens. Randomized prospective studies are needed.
Collapse
|
104
|
Jaramillo S, Krisam J, Le Cornet L, Kratzmann M, Baumann L, Sauer T, Crysandt M, Rank A, Behringer D, Teichmann L, Görner M, Trappe RU, Röllig C, Krause S, Hanoun M, Hopfer O, Held G, Buske S, Fransecky L, Kayser S, Schliemann C, Schaefer-Eckart K, Al-Fareh Y, Schubert J, Geer T, Kaufmann M, Brecht A, Niemann D, Kieser M, Bornhäuser M, Platzbecker U, Serve H, Baldus CD, Müller-Tidow C, Schlenk RF. Rationale and design of the 2 by 2 factorial design GnG-trial: a randomized phase-III study to compare two schedules of gemtuzumab ozogamicin as adjunct to intensive induction therapy and to compare double-blinded intensive postremission therapy with or without glasdegib in older patients with newly diagnosed AML. Trials 2021; 22:765. [PMID: 34732236 PMCID: PMC8564967 DOI: 10.1186/s13063-021-05703-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/01/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Overall survival remains poor in older patients with acute myeloid leukemia (AML) with less than 10% being alive after 5 years. In recent studies, a significant improvement in event-free, relapse-free and overall survival was shown by adding gemtuzumab ozogamicin (GO), a humanized antibody-drug conjugate directed against CD33, to intensive induction therapy once or in a sequential dosing schedule. Glasdegib, the small-molecule inhibitor of smoothened (SMO), also showed improved overall survival in patients not eligible for intensive chemotherapy when combined with low-dose cytarabine compared to low-dose cytarabine alone. These findings warrant further investigations in the phase III GnG trial. METHODS/DESIGN This is a randomized phase III trial with measurable residual disease (MRD) after induction therapy and event-free survival (EFS) as primary endpoints. The two research questions are addressed in a 2 by 2 factorial design. Patients age 60 years and older are upfront randomized 1:1 in one of the two induction arms: GO administered to intensive induction therapy on days 1,4, and 7 versus GO administered once on day 1 (GO-147 versus GO-1), and double-blinded 1:1 in one of the subsequent treatment arms glasdegib vs. placebo as adjunct to consolidation therapy and as single-agent maintenance therapy for six months. Chemotherapy backbone for induction therapy consists of standard 7 + 3 schedule with cytarabine 200 mg/m2 continuously days 1 to 7, daunorubicin 60 mg/m2 days 1, 2, and 3 and high-dose cytarabine (1 g/m2, bi-daily, days 1, 2, and 3) for consolidation therapy. Addressing two primary endpoints, MRD-negativity after induction therapy and event-free survival (EFS), 252 evaluable patients are needed to reject each of the two null hypotheses at a two-sided significance level of 2.5% with a power of at least 85%. ETHICS AND DISSEMINATION Ethical approval and approvals from the local and federal competent authorities were granted. Trial results will be reported via peer-reviewed journals and presented at conferences and scientific meetings. TRIAL STATUS Protocol version: 1st version 20.10.2020, no amendments yet. Study initiation on February 16, 2021. First patient was recruited on April 1st. TRIAL REGISTRATION ClinicalTrials.gov NCT04093505 ; EudraCT 2019-003913-32. Registered on October 30, 2018.
Collapse
Affiliation(s)
- Sonia Jaramillo
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.
| | - Johannes Krisam
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Lucian Le Cornet
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Markus Kratzmann
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Lukas Baumann
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Tim Sauer
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Martina Crysandt
- Department of Medicine IV, Aachen University Hospital, Aachen, Germany
| | - Andreas Rank
- Department of Medicine II, Augsburg University Hospital, Augsburg, Germany
| | - Dirk Behringer
- Department of Hematology, Oncology and Palliative Medicine, Augusta Hospital Bochum, Bochum, Germany
| | - Lino Teichmann
- Department of Medicine and Polyclinic III, Bonn University Hospital, Bonn, Germany
| | - Martin Görner
- Department of Hematology, Oncology and Palliative Medicine, Community Hospital Bielefeld, Bielefeld, Germany
| | - Ralf-Ulrich Trappe
- Department of Medicine II, Prot. Diaconal Hospital Bremen, Bremen, Germany
| | - Christoph Röllig
- Department of Internal Medicine I, TU Dresden University Hospital, Dresden, Germany
| | - Stefan Krause
- Department of Medicine V, Erlangen University Hospital, Erlangen, Germany
| | - Maher Hanoun
- Department of Hematology, Essen University Hospital, Essen, Germany
| | - Olaf Hopfer
- Department of Medicine I, Hospital Frankfurt (Oder), Frankfurt (Oder), Germany
| | - Gerhard Held
- Department of Internal Medicine I, Westpfalz Hospital Kaiserslautern, Kaiserslautern, Germany
| | - Sebastian Buske
- Department of Medicine II, Community Hospital Kiel, Kiel, Germany
| | - Lars Fransecky
- Department of Internal Medicine II, Schleswig-Holstein University Hospital Kiel, Kiel, Germany
| | - Sabine Kayser
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany.,Department of Medicine I - Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | | | | | - Yousef Al-Fareh
- Department of Hematology and Oncology, St. Josef Brothers' Hospital Paderborn, Paderborn, Germany
| | - Jörg Schubert
- Department of Internal Medicine II, Elbland Hospital Riesa, Riesa, Germany
| | - Thomas Geer
- Department of Medicine II, Diaconal Hospital Schwäbisch-Hall, Schwäbisch Hall, Germany
| | - Martin Kaufmann
- Department of Hematology, Oncology and Palliative Medicine, Robert-Bosch Hospital Stuttgart, Stuttgart, Germany
| | - Arne Brecht
- Department of Internal Medicine II, Helios Dr. Horst Schmidt Hospital Wiesbaden, Wiesbaden, Germany
| | - Dirk Niemann
- Department of Internal Medicine, Hematology, Oncology and Palliative Medicine, Prot. Monastery Hospital St. Jakob Koblenz, Koblenz, Germany
| | - Meinhard Kieser
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Martin Bornhäuser
- Department of Internal Medicine I, TU Dresden University Hospital, Dresden, Germany
| | - Uwe Platzbecker
- Department of Medicine I - Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | - Hubert Serve
- Department of Hematology/Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Claudia D Baldus
- Department of Internal Medicine II, Schleswig-Holstein University Hospital Kiel, Kiel, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard F Schlenk
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
105
|
Kennedy VE, Olin RL. Haematopoietic stem-cell transplantation in older adults: geriatric assessment, donor considerations, and optimisation of care. LANCET HAEMATOLOGY 2021; 8:e853-e861. [PMID: 34624239 DOI: 10.1016/s2352-3026(21)00231-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
Haematopoietic stem-cell transplantation (HSCT) has seen substantial growth among older adults. Chronological age is no longer viewed as an absolute barrier to HSCT, and alternative methods for assessing pre-transplantation fitness are increasingly used. In this Series paper, we summarise the metrics for pre-transplantation risk assessment in older adults, including both traditional metrics and geriatric assessment, and the ability of these metrics to predict post-transplantation outcomes. We also discuss strategies to broaden the utility of geriatric assessment, including in chronologically younger HSCT candidates and to guide individualised pre-transplantation interventions. Finally, we discuss donor considerations in older adults, including use of older sibling donors, haploidentical donors, and emerging data for donor-associated clonal haematopoiesis of indeterminate potential.
Collapse
Affiliation(s)
- Vanessa E Kennedy
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Rebecca L Olin
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
106
|
Better the cure you know: why patients with AML ≥60 years of age should be offered early allogeneic stem cell transplantation. Blood Adv 2021; 6:1619-1622. [PMID: 34607349 PMCID: PMC8905700 DOI: 10.1182/bloodadvances.2021004829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
107
|
Short NJ, Kantarjian H. When Less Is More: Reevaluating the Role of Intensive Chemotherapy for Older Adults With Acute Myeloid Leukemia in the Modern Era. J Clin Oncol 2021; 39:3104-3108. [PMID: 34406793 PMCID: PMC8478366 DOI: 10.1200/jco.21.00960] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/29/2021] [Accepted: 07/30/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
108
|
Guidelines for Pediatric Unrelated Cord Blood Transplantation-Unique Considerations. Transplant Cell Ther 2021; 27:968-972. [PMID: 34571212 DOI: 10.1016/j.jtct.2021.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/19/2021] [Indexed: 11/24/2022]
Abstract
Cord blood (CB) is the stem cell source of choice for approximately 30% of pediatric patients undergoing hematopoietic cell transplantation. Cord blood is readily available and is a particularly appealing stem cell source for patients who lack appropriate HLA-matched related or unrelated donors. Pediatric cord blood transplant (CBT) recipients have low rates of disease relapse in the malignant setting and very low rates of chronic graft-versus-host disease (GVHD). In addition, CB has unique properties that make it the stem cell source of choice for some nonmalignant conditions such as metabolic disorders. This review provides evidence-based and experience-based pediatric-specific guidelines for CBT including considerations for infectious disease management, CB unit selection and infusion, conditioning regimen selection, and GVHD management. In addition, it covers unique bedside considerations for pediatric patients and CB banking. In concert with the other topic specific CB guidelines previously published in this series, it provides a comprehensive overview of the clinical management of pediatric CBT.
Collapse
|
109
|
Weigert N, Rowe JM, Lazarus HM, Salman MY. Consolidation in AML: Abundant opinion and much unknown. Blood Rev 2021; 51:100873. [PMID: 34483002 DOI: 10.1016/j.blre.2021.100873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/24/2021] [Accepted: 08/01/2021] [Indexed: 11/25/2022]
Abstract
Consolidation therapy forms the backbone of post-remission therapy for AML and is uniformly accepted as an integral part of therapy designed to achieve long-term survival. The need for post-remission therapy was initially described over four decades ago and has since undergone many variations in terms of dosage, number of cycles and intensity of therapy. There is much empiricism in the current understanding of consolidation therapy and much that has not been rigorously studied. This review will consider the many aspects of consolidation therapy, focusing on the number of cycles, differences between young and older adults, first and subsequent remission as well as therapy prior to an allogeneic transplant. Emphasis will be given to differentiate strategies that are clearly evidence-based from those that have been incorporated into standard of care while bypassing the need for rigorous data-driven approaches. Finally, consideration will be given to the current ability to assess the minimal measureable residual disease and the impact that this may have on therapeutic paradigms, including superseding many of the time-honored prognostic features.
Collapse
Affiliation(s)
- Nir Weigert
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Jacob M Rowe
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel; Department of Hematology and Bone Marrow Transplantation, Rambam Medical Center, Haifa, Israel; Technion, Israel Institute of Technology, Haifa, Israel.
| | | | | |
Collapse
|
110
|
Aitken MJL, Ravandi F, Patel KP, Short NJ. Prognostic and therapeutic implications of measurable residual disease in acute myeloid leukemia. J Hematol Oncol 2021; 14:137. [PMID: 34479626 PMCID: PMC8417965 DOI: 10.1186/s13045-021-01148-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/25/2021] [Indexed: 01/10/2023] Open
Abstract
Quantification of measurable residual disease (MRD) provides critical prognostic information in acute myeloid leukemia (AML). A variety of platforms exist for MRD detection, varying in their sensitivity and applicability to individual patients. MRD detected by quantitative polymerase chain reaction, multiparameter flow cytometry, or next-generation sequencing has prognostic implications in various subsets of AML and at various times throughout treatment. While it is overwhelmingly evident that minute levels of remnant disease confer increased risk of relapse and shortened survival, the therapeutic implications of MRD remain less clear. The use of MRD as a guide to selecting the most optimal post-remission therapy, including hematopoietic stem cell transplant or maintenance therapy with hypomethylating agents, small molecule inhibitors, or immunotherapy is an area of active investigation. In addition, whether there are sufficient data to use MRD negativity as a surrogate endpoint in clinical trial development is controversial. In this review, we will critically examine the methods used to detect MRD, its role as a prognostic biomarker, MRD-directed therapeutics, and its potential role as a study endpoint.
Collapse
Affiliation(s)
- Marisa J L Aitken
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,McGovern Medical School, UT Health Science Center-Houston, Houston, TX, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
111
|
The Role of Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Leukemia. J Clin Med 2021; 10:jcm10173790. [PMID: 34501237 PMCID: PMC8432223 DOI: 10.3390/jcm10173790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/08/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) offers potentially curative treatment for many children with high-risk or relapsed acute leukemia (AL), thanks to the combination of intense preparative radio/chemotherapy and the graft-versus-leukemia (GvL) effect. Over the years, progress in high-resolution donor typing, choice of conditioning regimen, graft-versus-host disease (GvHD) prophylaxis and supportive care measures have continuously improved overall transplant outcome, and recent successes using alternative donors have extended the potential application of allotransplantation to most patients. In addition, the importance of minimal residual disease (MRD) before and after transplantation is being increasingly clarified and MRD-directed interventions may be employed to further ameliorate leukemia-free survival after allogeneic HSCT. These advances have occurred in parallel with continuous refinements in chemotherapy protocols and the development of targeted therapies, which may redefine the indications for HSCT in the coming years. This review discusses the role of HSCT in childhood AL by analysing transplant indications in both acute lymphoblastic and acute myeloid leukemia, together with current and most promising strategies to further improve transplant outcome, including optimization of conditioning regimen and MRD-directed interventions.
Collapse
|
112
|
Loo S, Wei AH. Post-transplant maintenance therapy for MDS and AML: a bridge too far or the beginning of a new era? Leuk Lymphoma 2021; 62:3073-3077. [PMID: 34348073 DOI: 10.1080/10428194.2021.1961243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Sun Loo
- Department of Clinical Hematology, Alfred Hospital, Melbourne, VIC, Australia
| | - Andrew H Wei
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
113
|
Talami A, Bettelli F, Pioli V, Giusti D, Gilioli A, Colasante C, Galassi L, Giubbolini R, Catellani H, Donatelli F, Maffei R, Martinelli S, Barozzi P, Potenza L, Marasca R, Trenti T, Tagliafico E, Comoli P, Luppi M, Forghieri F. How to Improve Prognostication in Acute Myeloid Leukemia with CBFB-MYH11 Fusion Transcript: Focus on the Role of Molecular Measurable Residual Disease (MRD) Monitoring. Biomedicines 2021; 9:biomedicines9080953. [PMID: 34440157 PMCID: PMC8391269 DOI: 10.3390/biomedicines9080953] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) carrying inv(16)/t(16;16), resulting in fusion transcript CBFB-MYH11, belongs to the favorable-risk category. However, even if most patients obtain morphological complete remission after induction, approximately 30% of cases eventually relapse. While well-established clinical features and concomitant cytogenetic/molecular lesions have been recognized to be relevant to predict prognosis at disease onset, the independent prognostic impact of measurable residual disease (MRD) monitoring by quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR), mainly in predicting relapse, actually supersedes other prognostic factors. Although the ELN Working Party recently indicated that patients affected with CBFB-MYH11 AML should have MRD assessment at informative clinical timepoints, at least after two cycles of intensive chemotherapy and after the end of treatment, several controversies could be raised, especially on the frequency of subsequent serial monitoring, the most significant MRD thresholds (most commonly 0.1%) and on the best source to be analyzed, namely, bone marrow or peripheral blood samples. Moreover, persisting low-level MRD positivity at the end of treatment is relatively common and not predictive of relapse, provided that transcript levels remain stably below specific thresholds. Rising MRD levels suggestive of molecular relapse/progression should thus be confirmed in subsequent samples. Further prospective studies would be required to optimize post-remission monitoring and to define effective MRD-based therapeutic strategies.
Collapse
Affiliation(s)
- Annalisa Talami
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Francesca Bettelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Valeria Pioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Davide Giusti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Andrea Gilioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Corrado Colasante
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Laura Galassi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Rachele Giubbolini
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Hillary Catellani
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Francesca Donatelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Rossana Maffei
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Silvia Martinelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Patrizia Barozzi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Leonardo Potenza
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Roberto Marasca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Unità Sanitaria Locale, 41126 Modena, Italy;
| | - Enrico Tagliafico
- Center for Genome Research, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy;
| | - Patrizia Comoli
- Pediatric Hematology/Oncology Unit and Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, 27100 Pavia, Italy;
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
- Correspondence: (M.L.); (F.F.); Tel.: +39-059-4222447 (F.F.); Fax: +39-059-4222386 (F.F.)
| | - Fabio Forghieri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
- Correspondence: (M.L.); (F.F.); Tel.: +39-059-4222447 (F.F.); Fax: +39-059-4222386 (F.F.)
| |
Collapse
|
114
|
Paiva B, Vidriales MB, Sempere A, Tarín F, Colado E, Benavente C, Cedena MT, Sánchez J, Caballero-Velazquez T, Cordón L, Garces JJ, Simoes C, Martínez-Cuadrón D, Bernal T, Botella C, Grille S, Serrano J, Rodríguez-Medina C, Algarra L, Alonso-Domínguez JM, Amigo ML, Barrios M, García-Boyero R, Colorado M, Pérez-Oteyza J, Pérez-Encinas M, Costilla-Barriga L, Sayas MJ, Pérez O, González-Díaz M, Pérez-Simón JA, Martínez-López J, Sossa C, Orfao A, San Miguel JF, Sanz MÁ, Montesinos P. Impact of measurable residual disease by decentralized flow cytometry: a PETHEMA real-world study in 1076 patients with acute myeloid leukemia. Leukemia 2021; 35:2358-2370. [PMID: 33526859 DOI: 10.1038/s41375-021-01126-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/09/2020] [Accepted: 01/07/2021] [Indexed: 01/29/2023]
Abstract
The role of decentralized assessment of measurable residual disease (MRD) for risk stratification in acute myeloid leukemia (AML) remains largely unknown, and so it does which methodological aspects are critical to empower the evaluation of MRD with prognostic significance, particularly if using multiparameter flow cytometry (MFC). We analyzed 1076 AML patients in first remission after induction chemotherapy, in whom MRD was evaluated by MFC in local laboratories of 60 Hospitals participating in the PETHEMA registry. We also conducted a survey on technical aspects of MRD testing to determine the impact of methodological heterogeneity in the prognostic value of MFC. Our results confirmed the recommended cutoff of 0.1% to discriminate patients with significantly different cumulative-incidence of relapse (-CIR- HR:0.71, P < 0.001) and overall survival (HR: 0.73, P = 0.001), but uncovered the limited prognostic value of MFC based MRD in multivariate and recursive partitioning models including other clinical, genetic and treatment related factors. Virtually all aspects related with methodological, interpretation, and reporting of MFC based MRD testing impacted in its ability to discriminate patients with different CIR. Thus, this study demonstrated that "real-world" assessment of MRD using MFC is prognostic in patients at first remission, and urges greater standardization for improved risk-stratification toward clinical decisions in AML.
Collapse
Affiliation(s)
- Bruno Paiva
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - María-Belen Vidriales
- Department of Hematology, University Hospital of Salamanca (HUS/IBSAL), CIBERONC (CB16/12/002333) and Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Amparo Sempere
- Hospital Universitario y Politécnico La Fe, CIBER-ONC number CB16/12/00284, Valencia, Spain
| | - Fabián Tarín
- Hospital General Universitario de Alicante, Alicante, Spain
| | - Enrique Colado
- Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria y Universitario Oncológico del Principado de Asturias (ISPA / IUOPA), Oviedo, Spain
| | | | | | | | - Teresa Caballero-Velazquez
- Hopsital Universitario Virgen del Rocío, Instituto de Biomedicina (IBIS / CSIC / CIBERONC), Universidad de Sevilla, Sevilla, Spain
| | - Lourdes Cordón
- Hospital Universitario y Politécnico La Fe, CIBER-ONC number CB16/12/00284, Valencia, Spain
| | - Juan-Jose Garces
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - Catia Simoes
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - David Martínez-Cuadrón
- Hospital Universitario y Politécnico La Fe, CIBER-ONC number CB16/12/00284, Valencia, Spain
| | - Teresa Bernal
- Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria y Universitario Oncológico del Principado de Asturias (ISPA / IUOPA), Oviedo, Spain
| | - Carmen Botella
- Hospital General Universitario de Alicante, Alicante, Spain
| | - Sofia Grille
- Hospital de Clinicas. Montevideo, Uruguay, Spain
| | | | | | | | | | | | - Manuel Barrios
- Hospital Regional Universitario de Málaga, Malaga, Spain
| | | | | | | | | | | | | | - Olga Pérez
- Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Marcos González-Díaz
- Department of Hematology, University Hospital of Salamanca (HUS/IBSAL), CIBERONC (CB16/12/002333) and Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
| | - José A Pérez-Simón
- Hopsital Universitario Virgen del Rocío, Instituto de Biomedicina (IBIS / CSIC / CIBERONC), Universidad de Sevilla, Sevilla, Spain
| | | | | | - Alberto Orfao
- Cancer Research Center (IBMCC-CSIC/USAL-IBSAL); Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Salamanca, Spain.,(USAL) Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Salamanca, Spain.,CIBER-ONC number CB16/12/00400, Salamanca, Spain
| | - Jesús F San Miguel
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - Miguel-Ángel Sanz
- Hospital Universitario y Politécnico La Fe, CIBER-ONC number CB16/12/00284, Valencia, Spain
| | - Pau Montesinos
- Hospital Universitario y Politécnico La Fe, CIBER-ONC number CB16/12/00284, Valencia, Spain.
| | | |
Collapse
|
115
|
Guolo F, Di Grazia C, Minetto P, Raiola AM, Clavio M, Miglino M, Tedone E, Contini P, Mangerini R, Kunkl A, Colombo N, Pugliese G, Carminati E, Marcolin R, Passannante M, Bagnasco S, Galaverna F, Lamparelli T, Ballerini F, Cagnetta A, Cea M, Gobbi M, Bacigalupo A, Lemoli RM, Angelucci E. Pre-transplant minimal residual disease assessment and transplant-related factors predict the outcome of acute myeloid leukemia patients undergoing allogeneic stem cell transplantation. Eur J Haematol 2021; 107:573-582. [PMID: 34297437 DOI: 10.1111/ejh.13694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
We studied pretransplant minimal residual disease (MRD) in 224 patients (median age 44 years; range 17-65) with acute myeloid leukemia (AML) undergoing allogeneic stem cell transplant (HSCT) in complete remission. MRD was evaluated on marrow samples using multicolor flow cytometry and assessment of WT1 gene expression. Both methods showed a strong prognostic value and their combination allowed the identification of three groups of patients with different risk of relapse. In multivariate analysis, combined MRD was the only predictor of cumulative incidence of relapse, regardless of donor type, conditioning regimen, first or second CR at HSCT, HSCT year, and ELN risk group. Multivariate regression model showed that only negative combined MRD status (P < .001) and myeloablative conditioning (P = .004) were independently associated with better OS. Among MRD-positive patients, a reduced incidence of relapse was observed in patients receiving haplo transplant (P < .05) and in patients who showed grade II-IV aGVHD (P < .03). In patients with negative combined MRD, the intensity of conditioning regimen did not affect the overall favorable outcome. We suggest that pretransplant MRD evaluation combined with transplant-related factors can identify AML patients at higher risk for relapse and might help in defining the overall transplant strategy.
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
| | | | - 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
| | - Maurizio Miglino
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Paola Contini
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | | | | | - Girolamo Pugliese
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Enrico Carminati
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Riccardo Marcolin
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Monica Passannante
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Samuele Bagnasco
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Federica Galaverna
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Filippo Ballerini
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Antonia Cagnetta
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Michele Cea
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Marco Gobbi
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, 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
|
116
|
Wang Y, Zhou W, McReynolds LJ, Katki HA, Griffiths EA, Thota S, Machiela MJ, Yeager M, McCarthy P, Pasquini M, Wang J, Karaesmen E, Rizvi A, Preus L, Tang H, Wang Y, Pooler L, Sheng X, Haiman CA, Van Den Berg D, Spellman SR, Wang T, Kuxhausen M, Chanock SJ, Lee SJ, Hahn TE, Sucheston-Campbell LE, Gadalla SM. Prognostic impact of pre-transplant chromosomal aberrations in peripheral blood of patients undergoing unrelated donor hematopoietic cell transplant for acute myeloid leukemia. Sci Rep 2021; 11:15004. [PMID: 34294836 PMCID: PMC8298542 DOI: 10.1038/s41598-021-94539-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/30/2021] [Indexed: 11/24/2022] Open
Abstract
To improve risk stratification and treatment decisions for patients with acute myeloid leukemia (AML) undergoing hematopoietic cell transplantation (HCT). We used SNP-array data from the DISCOVeRY-BMT study to detect chromosomal aberrations in pre-HCT peripheral blood (collected 2–4 weeks before the administration of conditioning regimen) from 1974 AML patients who received HCT between 2000 and 2011. All aberrations detected in ≥ 10 patients were tested for their association with overall survival (OS), separately by remission status, using the Kaplan–Meier estimator. Cox regression models were used for multivariable analyses. Follow-up was through January 2019. We identified 701 unique chromosomal aberrations in 285 patients (7% of 1438 in complete remission (CR) and 36% of 536 not in CR). Copy-neutral loss-of-heterozygosity (CNLOH) in chr17p in CR patients (3-year OS = 20% vs. 50%, with and without chr17p CNLOH, p = 0.0002), and chr13q in patients not in CR (3-year OS = 4% vs. 26%, with and without chr13q CNLOH, p < 0.0001) are risk factors for poor survival. Models adjusted for clinical factors showed approximately three-fold excess risk of post-HCT mortality with chr17p CNLOH in CR patients (hazard ratio, HR = 3.39, 95% confidence interval CI 1.74–6.60, p = 0.0003), or chr13q CNLOH in patients not in CR (HR = 2.68, 95% CI 1.75–4.09, p < 0.0001). The observed mortality was mostly driven by post-HCT relapse (HR = 2.47, 95% CI 1.01–6.02, p = 0.047 for chr17p CNLOH in CR patients, and HR = 2.58, 95% CI 1.63–4.08, p < 0.0001 for chr13q CNLOH in patients not in CR. Pre-transplant CNLOH in chr13q or chr17p predicts risk of poor outcomes after unrelated donor HCT in AML patients. A large prospective study is warranted to validate the results and evaluate novel strategies to improve survival in those patients.
Collapse
Affiliation(s)
- Youjin Wang
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA
| | - Weiyin Zhou
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA.,Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD, USA
| | - Lisa J McReynolds
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA
| | - Hormuzd A Katki
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA
| | | | - Swapna Thota
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mitchell J Machiela
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA
| | - Meredith Yeager
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA.,Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD, USA
| | - Philip McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marcelo Pasquini
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Junke Wang
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Ezgi Karaesmen
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Abbas Rizvi
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Leah Preus
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Hancong Tang
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yiwen Wang
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Loreall Pooler
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xin Sheng
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - David Van Den Berg
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephen R Spellman
- Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA
| | - Tao Wang
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA.,Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michelle Kuxhausen
- Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA
| | - Stephen J Chanock
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA
| | - Stephanie J Lee
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Theresa E Hahn
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Shahinaz M Gadalla
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA.
| |
Collapse
|
117
|
Jaramillo S, Schlenk RF. Post-Induction Treatment for Acute Myeloid Leukemia: Something Change? Curr Oncol Rep 2021; 23:109. [PMID: 34272619 PMCID: PMC8285306 DOI: 10.1007/s11912-021-01092-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Until recently, improvement in terms of survival for patients with acute myeloid leukemia (AML) was achieved mostly in younger patients with dose intensification of conventional chemotherapy and a broadening use of allogeneic hematopoietic cell transplantation (allo-HCT) whereas the results remained dismal and very stable in patients older than 60 years. The current review highlights the recent developments in standard intensive post-remission chemotherapy, evidence for the use of recently approved agents, and discusses the relevance of measurable residual disease (MRD) measurement in treatment adaptation. RECENT FINDINGS Current approvals of midostaurin, venetoclax, gemtuzumab ozogamicin, VYXEOS, ivosidenib, enasidenib, glasdegib, and CC-486 have changed the structure, aim, and schedule of consolidation therapy, and new, well-tolerated agents are being evaluated as maintenance therapies. Furthermore, MRD assessment has been implemented to guide the duration and type of consolidation and maintenance therapy as well as indicate the optimal timing of allo-HCT. Novel therapies have changed the structure and perspective of post-remission therapy in AML for both young and elderly patients. In addition, MRD assessment could guide the type, duration, and intensity of consolidation and maintenance therapy.
Collapse
Affiliation(s)
- Sonia Jaramillo
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Richard F. Schlenk
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
- NCT-Trial Center, NCT Heidelberg, DKFZ and Heidelberg University Hospital, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| |
Collapse
|
118
|
Freeman SD, Craddock C. Selection of Conditioning Intensity for Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemia and Myelodysplasia - New Evidence Emerges. Transplant Cell Ther 2021; 27:443-445. [PMID: 34144790 DOI: 10.1016/j.jtct.2021.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Sylvie D Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom.
| | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom; Cancer Research UK Clinical Trials Unit, University of Birmingham, United Kingdom
| |
Collapse
|
119
|
Gagelmann N, Kröger N. Dose intensity for conditioning in allogeneic hematopoietic cell transplantation: can we recommend "when and for whom" in 2021? Haematologica 2021; 106:1794-1804. [PMID: 33730842 PMCID: PMC8252938 DOI: 10.3324/haematol.2020.268839] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
Allogeneic hematopoietic stem-cell transplantation is a potentially curative therapy for various hematologic diseases. An essential component of this procedure is the pre-transplant conditioning regimen, which should facilitate engraftment and reduce or eliminate tumor cells. The recognition of the substantial association of a graft-versus- tumor effect and the high toxicity of the commonly used conditioning regimen led to the introduction of more differentiated intensity strategies, with the aim of making hematopoietic stem-cell transplantation less toxic and safer, and thus more applicable to broader populations such as older or unfit patients. In general, prospective and retrospective studies suggest a correlation between increasing intensity and nonrelapse mortality and an inverse correlation with relapse incidence. In this review, we will summarize traditional and updated definitions for conditioning intensity strategies and the landscape of comparative prospective and retrospective studies, which may help to find the balance between the risk of non-relapse mortality and relapse. We will try to underscore the caveats regarding these definitions and analyses, by missing complex differences between intensity and toxicity as well as the broad influences of other factors in the transplantation procedure. We will summarize evidence regarding several confounders which may influence decisions when selecting the intensity of the conditioning regimen for any given patient, according to the individual risk of relapse and non-relapse mortality.
Collapse
Affiliation(s)
- Nico Gagelmann
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg.
| |
Collapse
|
120
|
Bernasconi P, Borsani O. Eradication of Measurable Residual Disease in AML: A Challenging Clinical Goal. Cancers (Basel) 2021; 13:3170. [PMID: 34202000 PMCID: PMC8268140 DOI: 10.3390/cancers13133170] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 12/18/2022] Open
Abstract
In non-promyelocytic (non-M3) AML measurable residual disease (MRD) detected by multi-parameter flow cytometry and molecular technologies, which are guided by Consensus-based guidelines and discover very low leukemic cell numbers far below the 5% threshold of morphological assessment, has emerged as the most relevant predictor of clinical outcome. Currently, it is well-established that MRD positivity after standard induction and consolidation chemotherapy, as well as during the period preceding an allogeneic hematopoietic stem cell transplant (allo-HSCT), portends to a significantly inferior relapse-free survival (RFS) and overall survival (OS). In addition, it has become absolutely clear that conversion from an MRD-positive to an MRD-negative state provides a favorable clinical outcome similar to that associated with early MRD negativity. Thus, the complete eradication of MRD, i.e., the clearance of the few leukemic stem cells-which, due to their chemo-radiotherapy resistance, might eventually be responsible of disease recurrence-has become an un-met clinical need in AML. Nowadays, this goal might potentially be achieved thanks to the development of novel innovative treatment strategies, including those targeting driver mutations, apoptosis, methylation patterns and leukemic proteins. The aim of this review is to analyze these strategies and to suggest any potential combination able to induce MRD negativity in the pre- and post-HSCT period.
Collapse
Affiliation(s)
- Paolo Bernasconi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
- Hematology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Oscar Borsani
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
121
|
Narlı Özdemir Z, Şahin U, Dalva K, Baltacı MA, Uslu A, Öztürk C, Cengiz Seval G, Toprak SK, Kurt Yüksel M, Topçuoğlu P, Arslan Ö, Özcan M, Beksaç M, İlhan O, Gürman G, Civriz Bozdağ S. Highlighting the Prognostic Importance of Measurable Residual Disease Among Acute Myeloid Leukemia Risk Factors. Turk J Haematol 2021; 38:111-118. [PMID: 33112099 PMCID: PMC8171203 DOI: 10.4274/tjh.galenos.2020.2020.0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: The optimal timing of measurable residual disease (MRD) evaluation in acute myeloid leukemia (AML) patients has not been well defined yet. We aimed to investigate the impact of MRD in pre- and post-allogeneic hematopoietic stem cell transplantation (AHSCT) periods on prognostic parameters. Materials and Methods: Seventy-seven AML patients who underwent AHSCT in complete morphological remission were included. MRD analyses were performed by 10-color MFC and 10-4 was defined as positive. Relapse risk and survival outcomes were assessed based on pre- and post-AHSCT MRD positivity. Results: The median age of the patients was 46 (range: 18-71) years, and 41 (53.2%) were male while 36 (46.8%) were female. The median follow-up after AHSCT was 12.2 months (range: 0.2-73.0). The 2-year overall survival (OS) in the entire cohort was 37.0%, with a significant difference between patients who were MRD-negative and MRD-positive before AHSCT, estimated as 63.0% versus 16.0%, respectively (p=0.005). MRD positivity at +28 days after AHSCT was also associated with significantly inferior 2-year OS when compared to MRD negativity (p=0.03). The risk of relapse at 1 year was 2.4 times higher (95% confidence interval: 1.1-5.6; p=0.04) in the pre-AHSCT MRD-positive group when compared to the MRD-negative group regardless of other transplant-related factors, including pre-AHSCT disease status (i.e., complete remission 1 and 2). Event-free survival (EFS) was significantly shorter in patients who were pre-AHSCT MRD-positive (p=0.016). Post-AHSCT MRD positivity was also related to an increased relapse risk. OS and EFS were significantly inferior among MRD-positive patients at +28 days after AHSCT (p=0.03 and p=0.019). Conclusion: Our results indicate the importance of MRD before and after AHSCT independently of other factors.
Collapse
Affiliation(s)
| | - Uğur Şahin
- Medicana International Ankara Hospital, Clinic of Hematology, Ankara, Turkey
| | - Klara Dalva
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Mehmet Akif Baltacı
- Ankara University Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey
| | - Atilla Uslu
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Cemaleddin Öztürk
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | | | - Selami Koçak Toprak
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Meltem Kurt Yüksel
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Pervin Topçuoğlu
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Önder Arslan
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Muhit Özcan
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Meral Beksaç
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Osman İlhan
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Günhan Gürman
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Sinem Civriz Bozdağ
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| |
Collapse
|
122
|
Azacitidine maintenance after allogeneic hematopoietic cell transplantation for MDS and AML. Blood Adv 2021; 5:1757-1759. [PMID: 33755090 DOI: 10.1182/bloodadvances.2020003839] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/09/2020] [Indexed: 12/23/2022] Open
|
123
|
Measurable residual disease in elderly acute myeloid leukemia: results from the PETHEMA-FLUGAZA phase 3 clinical trial. Blood Adv 2021; 5:760-770. [PMID: 33560390 DOI: 10.1182/bloodadvances.2020003195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
The value of measurable residual disease (MRD) in elderly patients with acute myeloid leukemia (AML) is inconsistent between those treated with intensive vs hypomethylating drugs, and unknown after semi-intensive therapy. We investigated the role of MRD in refining complete remission (CR) and treatment duration in the phase 3 FLUGAZA clinical trial, which randomized 283 elderly AML patients to induction and consolidation with fludarabine plus cytarabine (FLUGA) vs 5-azacitidine. After consolidation, patients continued treatment if MRD was ≥0.01% or stopped if MRD was <0.01%, as assessed by multidimensional flow cytometry (MFC). On multivariate analysis including genetic risk and treatment arm, MRD status in patients achieving CR (N = 72) was the only independent prognostic factor for relapse-free survival (RFS) (HR, 3.45; P = .002). Achieving undetectable MRD significantly improved RFS of patients with adverse genetics (HR, 0.32; P = .013). Longer overall survival was observed in patients with undetectable MRD after induction though not after consolidation. Although leukemic cells from most patients displayed phenotypic aberrancies vs their normal counterpart (N = 259 of 265), CD34 progenitors from cases with undetectable MRD by MFC carried extensive genetic abnormalities identified by whole-exome sequencing. Interestingly, the number of genetic alterations significantly increased from diagnosis to MRD stages in patients treated with FLUGA vs 5-azacitidine (2.2-fold vs 1.1-fold; P = .001). This study supports MRD assessment to refine CR after semi-intensive therapy or hypomethylating agents, but unveils that improved sensitivity is warranted to individualize treatment and prolong survival of elderly AML patients achieving undetectable MRD.
Collapse
|
124
|
Prognostic impact of complete remission with MRD negativity in patients with relapsed or refractory AML. Blood Adv 2021; 4:6117-6126. [PMID: 33351107 DOI: 10.1182/bloodadvances.2020002811] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
In relapsed/refractory acute myeloid leukemia (AML), the prognostic impact of complete remission (CR) and measurable residual disease (MRD) negativity is not well established. We retrospectively analyzed 141 patients with relapsed/refractory AML who received first salvage therapy and had MRD assessed by multiparameter flow cytometry at the time of response. Patients who achieved CR with full hematologic recovery as best response vs those with incomplete hematology recovery had lower cumulative incidence of relapse (P = .01) and better relapse-free survival (P = .004) but not overall survival (P = .15); a similar trend was observed in patients who achieved MRD negativity vs those who were MRD positive (P = .01, P = .05, and P = .21, respectively). By multivariate analysis, CR and MRD negativity were each independently associated with lower cumulative incidence of relapse (P = .001 and P = .003, respectively) and better relapse-free survival (P < .001 and P = .02) but not overall survival. Patients who achieved CR with MRD negativity had the lowest rates of relapse and best survival (2-year overall survival rate, 37%), which was driven largely by lower rates of early relapse and an increased ability in this group to undergo hematopoietic stem cell transplantation (HSCT); however, post-HSCT outcomes were similar regardless of response to salvage chemotherapy. Overall, in patients with relapsed/refractory AML, CR with MRD negativity was associated with the best outcomes, supporting it as the optimal response in this setting.
Collapse
|
125
|
Measurable residual disease status and outcome of transplant in acute myeloid leukemia in second complete remission: a study by the acute leukemia working party of the EBMT. Blood Cancer J 2021; 11:88. [PMID: 33980810 PMCID: PMC8116335 DOI: 10.1038/s41408-021-00479-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 11/07/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Measurable residual disease (MRD) prior to hematopoietic cell transplant (HCT) for acute myeloid leukemia (AML) in first complete morphological remission (CR1) is an independent predictor of outcome, but few studies address CR2. This analysis by the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation registry assessed HCT outcomes by declared MRD status in a cohort of 1042 adult patients with AML CR2 at HCT. Patients were transplanted 2006–2016 from human leukocyte antigen (HLA) matched siblings (n = 719) or HLA 10/10 matched unrelated donors (n = 293). Conditioning was myeloablative (n = 610) or reduced-intensity (n = 432) and 566 patients (54%) had in-vivo T cell depletion. At HCT, 749 patients (72%) were MRD negative (MRD NEG) and 293 (28%) were MRD positive (MRD POS). Time from diagnosis to HCT was longer in MRD NEG than MRD POS patients (18 vs. 16 months (P < 0.001). Two-year relapse rates were 24% (95% CI, 21–28) and 40% (95% CI, 34–46) in MRD NEG and MRD POS groups (P < 0.001), respectively. Leukemia-free survival (LFS) was 57% (53–61) and 46% (40–52%), respectively (P = 0.001), but there was no difference in terms of overall survival. Prognostic factors for relapse and LFS were MRD NEG status, good risk cytogenetics, and longer time from diagnosis to HCT. In-vivo T cell depletion predicted relapse.
Collapse
|
126
|
Heuser M, Heida B, Büttner K, Wienecke CP, Teich K, Funke C, Brandes M, Klement P, Liebich A, Wichmann M, Neziri B, Chaturvedi A, Kloos A, Mintzas K, Gaidzik VI, Paschka P, Bullinger L, Fiedler W, Heim A, Puppe W, Krauter J, Döhner K, Döhner H, Ganser A, Stadler M, Hambach L, Gabdoulline R, Thol F. Posttransplantation MRD monitoring in patients with AML by next-generation sequencing using DTA and non-DTA mutations. Blood Adv 2021; 5:2294-2304. [PMID: 33929500 PMCID: PMC8114555 DOI: 10.1182/bloodadvances.2021004367] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/15/2021] [Indexed: 01/22/2023] Open
Abstract
Next-generation sequencing (NGS)-based measurable residual disease (MRD) monitoring in patients with acute myeloid leukemia (AML) is widely applicable and prognostic prior to allogeneic hematopoietic cell transplantation (alloHCT). We evaluated the prognostic role of clonal hematopoiesis-associated DNMT3A, TET2, and ASXL1 (DTA) and non-DTA mutations for MRD monitoring post-alloHCT to refine MRD marker selection. Of 154 patients with AML, 138 (90%) had at least one mutation at diagnosis, which were retrospectively monitored by amplicon-based error-corrected NGS on day 90 and/or day 180 post-alloHCT. MRD was detected in 34 patients on day 90 and/or day 180 (25%). The rate of MRD positivity was similar when DTA and non-DTA mutations were considered separately (17.6% vs 19.8%). DTA mutations had no prognostic impact on cumulative incidence of relapse, relapse-free survival, or overall survival in our study and were removed from further analysis. In the remaining 131 patients with at least 1 non-DTA mutation, clinical and transplantation-associated characteristics were similarly distributed between MRD-positive and MRD-negative patients. In multivariate analysis, MRD positivity was an independent adverse predictor of cumulative incidence of relapse, relapse-free survival, and overall survival but not of nonrelapse mortality. The prognostic effect was independent of different cutoffs (above limit of detection, 0.1% and 1% variant allele frequency). MRD log-reduction between diagnosis and post-alloHCT assessment had no prognostic value. MRD status post-alloHCT had the strongest impact in patients who were MRD positive prior to alloHCT. In conclusion, non-DTA mutations are prognostic NGS-MRD markers post-alloHCT, whereas the prognostic role of DTA mutations in the posttransplant setting remains open.
Collapse
Affiliation(s)
- Michael Heuser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Bennet Heida
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Konstantin Büttner
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Clara Philine Wienecke
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Katrin Teich
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Carolin Funke
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Maximilian Brandes
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Piroska Klement
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Alessandro Liebich
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Martin Wichmann
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Blerina Neziri
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Anuhar Chaturvedi
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Arnold Kloos
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Konstantinos Mintzas
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Verena I Gaidzik
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Peter Paschka
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Walter Fiedler
- Department of Medicine II, Oncological Center, Hubertus Wald University Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Albert Heim
- Department of Virology, Hannover Medical School, Hannover, Germany; and
| | - Wolfram Puppe
- Department of Virology, Hannover Medical School, Hannover, Germany; and
| | - Jürgen Krauter
- Department of Hematology and Oncology, Klinikum Braunschweig, Braunschweig, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Michael Stadler
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Lothar Hambach
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Razif Gabdoulline
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
127
|
Doucette K, Karp J, Lai C. Advances in therapeutic options for newly diagnosed, high-risk AML patients. Ther Adv Hematol 2021; 12:20406207211001138. [PMID: 33995985 PMCID: PMC8111550 DOI: 10.1177/20406207211001138] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/18/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive malignancy characterized by clonal proliferation of neoplastic immature precursor cells. AML impacts older adults and has a poor prognosis. Despite recent advances in treatment, AML is complex, with both genetic and epigenetic aberrations in the malignant clone and elaborate interactions with its microenvironment. We are now able to stratify patients on the basis of specific clinical and molecular features in order to optimize individual treatment strategies. However, our understanding of the complex nature of these molecular abnormalities continues to expand the defining characteristics of high-risk mutations. In this review, we focus on genetic and microenvironmental factors in adverse risk AML that play critical roles in leukemogenesis, including those not described in an European LeukemiaNet adverse risk group, and describe therapies that are currently in the clinical arena, either approved or under development.
Collapse
Affiliation(s)
- Kimberley Doucette
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Judith Karp
- Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Catherine Lai
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, 3800 Reservoir Road, NW, Washington, DC 20007, USA
| |
Collapse
|
128
|
Gui G, Dillon LW, Hourigan CS. Measurable Residual Disease Before Reduced-Intensity Allogeneic Transplantation in Patients With Myeloid Malignancy. J Clin Oncol 2021; 39:2413-2415. [PMID: 33950704 DOI: 10.1200/jco.21.00255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Gege Gui
- Gege Gui, ScM, Laura W. Dillon, PhD, and ChristopherS. Hourigan, DM, DPhil, Laboratory of Myeloid Malignancy, Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Laura W Dillon
- Gege Gui, ScM, Laura W. Dillon, PhD, and ChristopherS. Hourigan, DM, DPhil, Laboratory of Myeloid Malignancy, Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Christopher S Hourigan
- Gege Gui, ScM, Laura W. Dillon, PhD, and ChristopherS. Hourigan, DM, DPhil, Laboratory of Myeloid Malignancy, Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
| |
Collapse
|
129
|
Wu X, Lu H, Pang T, Li X, Luo H, Tan H, Liu S. Association of minimal residual disease levels with clinical outcomes in patients with mantle cell lymphoma: A meta-analysis. Leuk Res 2021; 108:106605. [PMID: 34090063 DOI: 10.1016/j.leukres.2021.106605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 02/08/2023]
Abstract
Some studies have elucidated that Minimal residual disease (MRD) in patient with Mantle Cell Lymphoma (MCL) was a significant prognostic factor, with potential value in assessing overall survival (OS) and progression-free survival (PFS). However, most studies were widely varied in included population, sample sources and MRD detection time points. Some studies even have conflicting results. In view of this, a meta-analysis was performed to evaluate association of MRD levels with clinical outcomes in patients with MCL. We identified 7 included articles, which were published in recent 20 years. Then, we extracted or calculated hazard ratios (HRs) and their 95 % confidence intervals (CIs). Our results reveal that patients with MRD negativity have improved OS (HR = 0.63; 95 % CI: 0.50-0.79) and PFS (HR = 0.40, 95 % CI: 0.21-0.76), comparing with patients with MRD positivity. There are also consistent results in subgroups based on sample sources and MRD detection time points. Our study also demonstrates that MRD level is a strong prognostic factor of clinical outcomes. Thus, MRD is expected to be an effective clinical indicator for assessing prognosis and guide treatment decisions in MCL patients.
Collapse
Affiliation(s)
- Xue Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan Province, China
| | - Hongyu Lu
- School of Medical Technology, Chengdu University of TCM, Chengdu, 611137, Sichuan Province, China; Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Pang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan Province, China
| | - Xue Li
- Department of Laboratory Medicine, Hanyuan People's Hospital, Yuan, 25000, Sichuan, China
| | - Hongzhi Luo
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hong Tan
- Department of General Surgery, Chengdu Integrated TCM&Western Medicine Hospital (Chengdu First People's Hospital), No.18 Vientiane North Road, Hi-tech Zone, Chengdu, 610041, China.
| | - Shan Liu
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No.32 West Second Section First Ring Road, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
130
|
Loke J, Buka R, Craddock C. Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia: Who, When, and How? Front Immunol 2021; 12:659595. [PMID: 34012445 PMCID: PMC8126705 DOI: 10.3389/fimmu.2021.659595] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022] Open
Abstract
Although the majority of patients with acute myeloid leukemia (AML) treated with intensive chemotherapy achieve a complete remission (CR), many are destined to relapse if treated with intensive chemotherapy alone. Allogeneic stem cell transplant (allo-SCT) represents a pivotally important treatment strategy in fit adults with AML because of its augmented anti-leukemic activity consequent upon dose intensification and the genesis of a potent graft-versus-leukemia effect. Increased donor availability coupled with the advent of reduced intensity conditioning (RIC) regimens has dramatically increased transplant access and consequently allo-SCT is now a key component of the treatment algorithm in both patients with AML in first CR (CR1) and advanced disease. Although transplant related mortality has fallen steadily over recent decades there has been no real progress in reducing the risk of disease relapse which remains the major cause of transplant failure and represents a major area of unmet need. A number of therapeutic approaches with the potential to reduce disease relapse, including advances in induction chemotherapy, the development of novel conditioning regimens and the emergence of the concept of post-transplant maintenance, are currently under development. Furthermore, the use of genetics and measurable residual disease technology in disease assessment has improved the identification of patients who are likely to benefit from an allo-SCT which now represents an increasingly personalized therapy. Future progress in optimizing transplant outcome will be dependent on the successful delivery by the international transplant community of randomized prospective clinical trials which permit examination of current and future transplant therapies with the same degree of rigor as is routinely adopted for non-transplant therapies.
Collapse
Affiliation(s)
- Justin Loke
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Richard Buka
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
131
|
Yu J, Du Y, Ahmad S, Patel RD, Varela JC, Chang CC, Mori S. Comparison of Myeloablative versus Reduced-Intensity Conditioning Regimens in Allogeneic Stem Cell Transplantation Recipients with Acute Myelogenous Leukemia with Measurable Residual Disease-Negative Disease at the Time of Transplantation: A Retrospective Cohort Study. Transplant Cell Ther 2021; 27:663.e1-663.e6. [PMID: 33951497 DOI: 10.1016/j.jtct.2021.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
The ideal conditioning intensity in allogeneic hematopoietic stem cell transplantation (HSCT) is evolving. Previous prospective studies comparing myeloablative conditioning (MAC) versus reduced-intensity conditioning (RIC) regimens in adults with acute myelogenous leukemia (AML) have shown mixed results. In many of these studies, patients were not stratified based on measurable residual disease (MRD). We evaluated the effect of conditioning intensity on the outcomes of AML patients in complete remission (CR) with flow cytometry evidence of MRD negativity. A total of 135 patients age 20 to 75 years with AML in CR1 or CR2 and flow cytometry evidence of MRD negativity who underwent allogeneic HSCT at our center between 2011 and 2019 were evaluated. We compared overall survival (OS), relapse-free survival (RFS), nonrelapse mortality (NRM), relapse, and acute and chronic graft-versus-host disease (GVHD) in recipients of MAC (n = 89) and RIC (n = 46). Although the patients receiving RIC were older (62 versus 51 years; P < .0001), there were no statistically significant differences between the groups in terms of Eastern Cooperative Oncology Group and European Leukemia Network risk criteria and disease status (CR1 or CR2) at the time of transplantation. At a median follow-up of 24.6 months, no statistically significant differences in OS (hazard ratio [HR], 0.78; 95% confidence interval [CI] 0.42 to 1.42, P = .411) or RFS (HR, 1.004; 95% CI, 0.48 to 2.09, P = .99) were identified. The cumulative incidence of NRM (HR, 0.595; 95% CI, 0.24 to 1.48; P = .2644) and relapse (HR, 1.007; 95% CI, 0.45 to 2.23; P = .9872) was not different between the 2 groups. Grade II-IV and grade III-IV acute GVHD were more frequent in the MAC group (39.3% verses 19.9% [P = .018] and 19.3% versus 2.3% [P < .001], respectively), as was moderate/severe chronic GVHD (23.6% versus 15.8%; P = .038). Our data indicate that conditioning intensity did not appear to affect OS, RFS, NRM, and relapse risk in patients with MRD-negative AML as measured by flow cytometry. RIC resulted in less severe acute and chronic GVHD.
Collapse
Affiliation(s)
- James Yu
- Department of Internal Medicine, AdventHealth Orlando Hospital, Orlando, Florida
| | - Yuan Du
- Research Institute, AdventHealth Orlando Hospital, Orlando, Florida
| | - Sarfraz Ahmad
- Gynecologic Oncology Program, AdventHealth Orlando Hospital, Orlando, Florida
| | - Rushang D Patel
- Blood and Marrow Transplant Center, AdventHealth Orlando Hospital, Orlando, Florida
| | - Juan Carlos Varela
- Blood and Marrow Transplant Center, AdventHealth Orlando Hospital, Orlando, Florida
| | - Chung-Che Chang
- Department of Pathology and Laboratory Medicine, AdventHealth Orlando Hospital, Orlando, Florida
| | - Shahram Mori
- Blood and Marrow Transplant Center, AdventHealth Orlando Hospital, Orlando, Florida.
| |
Collapse
|
132
|
Arora S, Pushpam D, Tiwari A, Choudhary P, Chopra A, Gupta R, Kumar R, Bakhshi S. Allogeneic hematopoietic stem cell transplant in pediatric acute myeloid leukemia: Lessons learnt from a tertiary care center in India. Pediatr Transplant 2021; 25:e13918. [PMID: 33142026 DOI: 10.1111/petr.13918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/09/2020] [Accepted: 10/18/2020] [Indexed: 01/10/2023]
Abstract
There is paucity of data on outcomes of MSD-HSCT in children with relapsed or high-risk AML from developing countries, which have unique challenges including adverse host factors and resource constraints. We retrospectively reviewed records of children (age ≤ 18 years) who underwent MSD-HSCT for AML at our center from 2009 to 2019 to evaluate clinical outcome and its predictors using Cox proportional hazards model. There were 46 children (36 boys and 10 girls) with mean age 10.7 ± 4.8 years. Indication for HSCT was relapsed AML in CR2 (n = 37), primary refractory (n = 3), or relapsed refractory disease (n = 3); high-risk (n = 1) or secondary (n = 2) AML in CR1. Five-year EFS and OS were 33.3 ± 7.2% and 36.3 ± 7.6%, respectively. On multivariate analysis, CR1 duration less than 12 months, presence of active disease at transplant, and use of bone marrow stem cell graft were associated with poorer EFS and OS. There was one (2.2%) TRM, while disease relapse occurred in 20/40 patients who underwent HSCT in remission. Though the 5-year EFS and OS were inferior to results reported from high-income countries, relapse (and not TRM) was the major cause of treatment failure. A well-sustained CR1, achievement of disease remission, and use of peripheral blood allograft seem imperative to a successful transplant. Targeted therapy along with HSCT may be the option for those with early relapse.
Collapse
Affiliation(s)
- Shalabh Arora
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Deepam Pushpam
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Akash Tiwari
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | | | - Anita Chopra
- Unit of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Ritu Gupta
- Unit of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Rajive Kumar
- Unit of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| |
Collapse
|
133
|
Loke J, Vyas H, Craddock C. Optimizing Transplant Approaches and Post-Transplant Strategies for Patients With Acute Myeloid Leukemia. Front Oncol 2021; 11:666091. [PMID: 33937080 PMCID: PMC8083129 DOI: 10.3389/fonc.2021.666091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Acute Myeloid Leukemia (AML) is the commonest indication for allogeneic stem cell transplantation (allo-SCT) worldwide. The increasingly important role of allo-SCT in the management of AML has been underpinned by two important advances. Firstly, improvements in disease risk stratification utilizing genetic and Measurable Residual Disease (MRD) technologies permit ever more accurate identification of allo-mandatory patients who are at high risk of relapse if treated by chemotherapy alone. Secondly, increased donor availability coupled with the advent of reduced intensity conditioning (RIC) regimens has substantially expanded transplant access for patients with high risk AML In patients allografted for AML disease relapse continues to represent the commonest cause of transplant failure and the development of novel strategies with the potential to reduce disease recurrence represents a major unmet need.
Collapse
Affiliation(s)
- Justin Loke
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom.,Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Hrushikesh Vyas
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom.,Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom.,Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
134
|
Walter RB, Ofran Y, Wierzbowska A, Ravandi F, Hourigan CS, Ngai LL, Venditti A, Buccisano F, Ossenkoppele GJ, Roboz GJ. Measurable residual disease as a biomarker in acute myeloid leukemia: theoretical and practical considerations. Leukemia 2021; 35:1529-1538. [PMID: 33758317 DOI: 10.1038/s41375-021-01230-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/19/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022]
Abstract
Several methodologies that rely on the detection of immunophenotypic or molecular abnormalities of the neoplastic cells are now available to quantify measurable ("minimal") residual disease (MRD) in acute myeloid leukemia (AML). Although the perfect MRD test does not (yet) exist, the strong association between MRD and adverse patient outcomes has provided the impetus to use measures of MRD as biomarker in the routine care of AML patients and during clinical trials. MRD test results may inform the selection of postremission therapy in some patients but evidence supporting the use of MRD as predictive biomarker is still limited. Several retrospective studies have shown that conversion from undetectable to detectable MRD or increasing MRD over time is associated with overt disease recurrence, and MRD testing may therefore be valuable as a monitoring biomarker for early detection of relapse. Interpreting serial MRD data is complex, with open questions regarding the optimal timing and frequency of testing, as well as the identification of test-specific thresholds to define relapse. Importantly, it is unknown whether intervening at the time of MRD detection, rather than at overt disease recurrence, improves outcomes. Finally, using MRD as a surrogate efficacy-response biomarker to accelerate drug development/approval has already been accepted by regulatory authorities in other diseases and is of great interest as a potential strategy in AML. While the prognostic value of MRD in AML is well established, data from prospective clinical trials confirming that treatment effects on MRD directly relate to clinical outcomes are needed to further establish the role of MRD as a surrogate endpoint in AML.
Collapse
Affiliation(s)
- Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA. .,Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA. .,Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA. .,Department of Epidemiology, University of Washington, Seattle, WA, USA.
| | - Yishai Ofran
- Department of Hematology, Sharee Zedeq Medical Center, Jerusalem, Israel
| | | | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lok Lam Ngai
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriano Venditti
- Department of Biomedicine and Prevention, Hematology, University Tor Vergata, Rome, Italy
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, Hematology, University Tor Vergata, Rome, Italy
| | - Gert J Ossenkoppele
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Gail J Roboz
- Hematology and Oncology, Weill Cornell Medicine and NewYork-Presbyterian Hospital, Cornell University, New York, NY, USA
| |
Collapse
|
135
|
Hochman MJ, Othus M, Walter RB, Shaw C, Gardner K, Percival MEM, Halpern AB, Hendrie PC, Sandmaier BM, Estey EH. Effect of post-treatment MRD status on subsequent outcomes according to chemotherapy intensity in acute myeloid leukemia (AML). Leuk Lymphoma 2021; 62:1532-1535. [PMID: 33752557 DOI: 10.1080/10428194.2021.1885658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Michael J Hochman
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| | - Megan Othus
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Roland B Walter
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Carole Shaw
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kelda Gardner
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| | - Mary-Elizabeth M Percival
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Anna B Halpern
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Paul C Hendrie
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| | - Brenda M Sandmaier
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Elihu H Estey
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
136
|
Rosso A, Juliusson G, Lorenz F, Lehmann S, Derolf Å, Deneberg S, Jädersten M, Antunovic P, Cammenga J, Möllgård L, Wennström L, Ölander E, Ehinger M, Fogelstrand L, Höglund M, Lazarevic VL. Is there an impact of measurable residual disease as assessed by multiparameter flow cytometry on survival of AML patients treated in clinical practice? A population-based study. Leuk Lymphoma 2021; 62:1973-1981. [PMID: 33719843 DOI: 10.1080/10428194.2021.1889539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The Swedish national guidelines for treatment of acute myeloid leukemia (AML) recommend analysis of measurable residual disease (MRD) by multiparameter flow cytometry (MFC) in bone marrow in the routine clinical setting. The Swedish AML registry contains such MRD data in AML patients diagnosed 2011-2019. Of 327 patients with AML (non-APL) with MRD-results reported in complete remission after two courses of intensive chemotherapy 229 were MRD-negative (70%), as defined by <0.1% cells with leukemia-associated immunophenotype in the bone marrow. MRD-results were reported to clinicians in real time. Multivariate statistical analysis adjusted for known established risk factors did not indicate an association between MFC-MRD and overall survival (HR: 1.00 [95% CI 0.61, 1.63]) with a median follow-up of 2.7 years. Knowledge of the importance of MRD status by clinicians and individualized decisions could have ameliorated the effects of MRD as an independent prognostic factor of overall survival.
Collapse
Affiliation(s)
- Aldana Rosso
- Department of Clinical Sciences, Division of Geriatric Medicine, Lund University, Lund, Sweden.,Diagnostic Radiology, Department of Translational Medicine, Lund University, Lund, Sweden
| | - Gunnar Juliusson
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden.,Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Fryderyk Lorenz
- Department of Oncology and Hematology, Umeå University Hospital, Umeå, Sweden
| | - Sören Lehmann
- Department of Medical Sciences, Division of Hematology, Uppsala University Hospital, Uppsala, Sweden
| | - Åsa Derolf
- Department of Hematology, Karolinska University Hospital, Huddinge, Sweden
| | - Stefan Deneberg
- Department of Hematology, Karolinska University Hospital, Huddinge, Sweden
| | - Martin Jädersten
- Department of Hematology, Karolinska University Hospital, Huddinge, Sweden
| | - Petar Antunovic
- Department of Hematology, Linköping University Hospital, Linköping, Sweden
| | - Jörg Cammenga
- Department of Hematology, Linköping University Hospital, Linköping, Sweden
| | - Lars Möllgård
- Department of Hematology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lovisa Wennström
- Department of Hematology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Emma Ölander
- Department of Hematology, Sundsvall Hospital, Sundsvall, Sweden
| | - Mats Ehinger
- Department of Clinical Sciences, Pathology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Linda Fogelstrand
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Martin Höglund
- Department of Medical Sciences, Division of Hematology, Uppsala University Hospital, Uppsala, Sweden
| | - Vladimir Lj Lazarevic
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden.,Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
137
|
Shouval R, Fein JA, Labopin M, Cho C, Bazarbachi A, Baron F, Bug G, Ciceri F, Corbacioglu S, Galimard JE, Giebel S, Gilleece MH, Giralt S, Jakubowski A, Montoto S, O'Reilly RJ, Papadopoulos EB, Peric Z, Ruggeri A, Sanz J, Sauter CS, Savani BN, Schmid C, Spyridonidis A, Tamari R, Versluis J, Yakoub-Agha I, Perales MA, Mohty M, Nagler A. Development and validation of a disease risk stratification system for patients with haematological malignancies: a retrospective cohort study of the European Society for Blood and Marrow Transplantation registry. LANCET HAEMATOLOGY 2021; 8:e205-e215. [PMID: 33636142 DOI: 10.1016/s2352-3026(20)30394-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diagnosis and remission status at the time of allogeneic haematopoietic stem-cell transplantation (HSCT) are the principal determinants of overall survival following transplantation. We sought to develop a contemporary disease-risk stratification system (DRSS) that accounts for heterogeneous transplantation indications. METHODS In this retrospective cohort study we included 55 histology and remission status combinations across haematological malignancies, including acute leukaemia, lymphoma, multiple myeloma, and myeloproliferative and myelodysplastic disorders. A total of 47 265 adult patients (aged ≥18 years) who received an allogeneic HSCT between Jan 1, 2012, and Dec 31, 2016, and were reported to the European Society for Blood and Marrow Transplantation registry were included. We divided EBMT patients into derivation (n=25 534), tuning (n=18 365), and geographical validation (n=3366) cohorts. Disease combinations were ranked in a multivariable Cox regression for overall survival in the derivation cohort, cutoff for risk groups were evaluated for the tuning cohort, and the selected system was tested on the geographical validation cohort. An independent single-centre US cohort of 660 patients transplanted between Jan 1, 2010, and Dec 31, 2015 was used to externally validate the results. FINDINGS The DRSS model stratified patients in the derivation cohort (median follow-up was 2·1 years [IQR 1·0-3·2]) into five risk groups with increasing mortality risk: low risk (reference group), intermediate-1 (hazard ratio for overall survival 1·26 [95% CI 1·17-1·36], p<0·0001), intermediate-2 (1·53 [1·42-1·66], p<0·0001), high (2·03 [1·86-2·22], p<0·0001), and very high (2·87 [2·63-3·13], p<0·0001). DRSS levels were also associated with a stepwise increase in risk across the tuning and geographical validation cohort. In the external validation cohort (median follow-up was 5·7 years [IQR 4·5-7·1]), the DRSS scheme separated patients into 4 risk groups associated with increasing risk of mortality: intermediate-2 risk (hazard ratio [HR] 1·34 [95% CI 1·04-1·74], p=0·025), high risk (HR 2·03 [95% CI 1·39-2·95], p=0·00023) and very-high risk (HR 2·26 [95% CI 1·62-3·15], p<0·0001) patients compared with the low risk and intermediate-1 risk group (reference group). Across all cohorts, between 64% and 65% of patients were categorised as having intermediate-risk disease by a previous prognostic system (ie, the disease-risk index [DRI]). The DRSS reclassified these intermediate-risk DRI patients, with 855 (6%) low risk, 7111 (51%) intermediate-1 risk, 5700 (41%) intermediate-2 risk, and 375 (3%) high risk or very high risk of 14 041 patients in a subanalysis combining the tuning and internal geographic validation cohorts. The DRI projected 2-year overall survival was 62·1% (95% CI 61·2-62·9) for these 14 041 patients, while the DRSS reclassified them into finer prognostic groups with overall survival ranging from 45·7% (37·4-54·0; very high risk patients) to 73·1% (70·1-76·2; low risk patients). INTERPRETATION The DRSS is a novel risk stratification tool including disease features related to histology, genetic profile, and treatment response. The model should serve as a benchmark for future studies. This system facilitates the interpretation and analysis of studies with heterogeneous cohorts, promoting trial-design with more inclusive populations. FUNDING The Varda and Boaz Dotan Research Center for Hemato-Oncology Research, Tel Aviv University.
Collapse
Affiliation(s)
- Roni Shouval
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hematology Division, Chaim Sheba Medical Center, Tel Aviv University, Ramat Gan, Israel.
| | - Joshua A Fein
- Internal Medicine, University of Connecticut, Farmington, CT, USA
| | - Myriam Labopin
- The European Society for Blood and Marrow Transplantation Paris Study Office, Paris, France
| | - Christina Cho
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ali Bazarbachi
- Department of Internal Medicine, Bone Marrow Transplantation Program, American University of Beirut, Beirut, Lebanon
| | - Frédéric Baron
- Division of Haematology, University of Liège, Liège, Belgium
| | - Gesine Bug
- Goethe-Universitat Frankfurt am Main, Frankfurt, Germany
| | - Fabio Ciceri
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Selim Corbacioglu
- Department of Pediatric Hematology, University Hospital Regensburg, Regensburg, Germany
| | | | - Sebastian Giebel
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | | | - Sergio Giralt
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ann Jakubowski
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Silvia Montoto
- Department of Haemato-oncology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Richard J O'Reilly
- Pediatric Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Esperanza B Papadopoulos
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zinaida Peric
- University Hospital Centre Zagreb, Zagreb School of Medicine, Zagreb, Croatia
| | | | - Jaime Sanz
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Craig S Sauter
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bipin N Savani
- Division of Hematology and Oncology, Vanderbilt University, Nashville, TN, USA
| | | | | | - Roni Tamari
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Miguel Angel Perales
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohamad Mohty
- INSERM UMRs 938, Paris, France; Service d'Hématologie Clinique et de Thérapie Cellulaire, Hospital Saint Antoine, Paris, France
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Aviv University, Ramat Gan, Israel
| |
Collapse
|
138
|
Buccisano F, Palmieri R, Piciocchi A, Maurillo L, Del Principe MI, Paterno G, Soddu S, Cerretti R, De Angelis G, Mariotti B, Irno Consalvo MA, Conti C, Fraboni D, Divona M, Ottone T, Lavorgna S, Panetta P, Voso MT, Arcese W, Venditti A. Use of Measurable Residual Disease to Evolve Transplant Policy in Acute Myeloid Leukemia: A 20-Year Monocentric Observation. Cancers (Basel) 2021; 13:1083. [PMID: 33802502 PMCID: PMC7959451 DOI: 10.3390/cancers13051083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/11/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Measurable residual disease (MRD) is increasingly employed as a biomarker of quality of complete remission (CR) in intensively treated acute myeloid leukemia (AML) patients. We evaluated if a MRD-driven transplant policy improved outcome as compared to a policy solely relying on a familiar donor availability. High-risk patients (adverse karyotype, FLT3-ITD) received allogeneic hematopoietic cell transplant (alloHCT) whereas for intermediate and low risk ones (CBF-AML and NPM1-mutated), alloHCT or autologous SCT was delivered depending on the post-consolidation measurable residual disease (MRD) status, as assessed by flow cytometry. For comparison, we analyzed a matched historical cohort of patients in whom alloHCT was delivered based on the sole availability of a matched sibling donor. Ten-years overall and disease-free survival were longer in the MRD-driven cohort as compared to the historical cohort (47.7% vs. 28.7%, p = 0.012 and 42.0% vs. 19.5%, p = 0.0003). The favorable impact of this MRD-driven strategy was evident for the intermediate-risk category, particularly for MRD positive patients. In the low-risk category, the significantly lower CIR of the MRD-driven cohort did not translate into a survival advantage. In conclusion, a MRD-driven transplant allocation may play a better role than the one based on the simple donor availability. This approach determines a superior outcome of intermediate-risk patients whereat in low-risk ones a careful evaluation is needed for transplant allocation.
Collapse
Affiliation(s)
- Francesco Buccisano
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Raffaele Palmieri
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | | | - Luca Maurillo
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Maria Ilaria Del Principe
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Giovangiacinto Paterno
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Stefano Soddu
- Centro Dati Fondazione GIMEMA, 00100 Rome, Italy; (A.P.); (S.S.)
| | - Raffaella Cerretti
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
- Rome Transplant Network, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Gottardo De Angelis
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
- Rome Transplant Network, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Benedetta Mariotti
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
- Rome Transplant Network, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Maria Antonietta Irno Consalvo
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Consuelo Conti
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Daniela Fraboni
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Mariadomenica Divona
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Serena Lavorgna
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Paola Panetta
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - William Arcese
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
- Rome Transplant Network, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Adriano Venditti
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| |
Collapse
|
139
|
Autologous hematopoietic cell transplantation following high-dose cytarabine consolidation for core-binding factor-acute myeloid leukemia in first complete remission: a phase 2 prospective trial. Int J Hematol 2021; 113:851-860. [PMID: 33655416 DOI: 10.1007/s12185-021-03099-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
Core-binding factor (CBF)-acute myeloid leukemia (AML) generally have a favorable prognosis. However, approximately 50% of patients experience disease relapse during or after post-remission therapy. Retrospective studies on autologous hematopoietic cell transplantation (AHCT) have shown improved survival with decreased relapse rate in CBF-AML. In this prospective study, we evaluate the outcomes of AHCT following high-dose cytarabine (HiDAC) consolidation in patients with CBF-AML in first complete remission (CR). Adult patients with CBF-AML achieving first CR after induction chemotherapy were eligible for the study. High-dose chemotherapy before AHCT included intravenous busulfan (3.2 mg/kg/day, days - 7 to - 5) and etoposide (400 mg/m2/day, days - 3 to - 2). Twenty-nine patients, 17 with t(8;21) and 12 with inv(16), underwent AHCT following 2 or 3 courses of HiDAC consolidation. The estimated 5-year overall and disease-free survival rates were between 89.0% and 82.5%, respectively. The cumulative incidences of relapse and non-relapse mortality were between 17.5% and 0%, respectively. Presence of measurable residual disease (MRD) before AHCT and KIT mutation were significantly associated with relapse after transplantation. In conclusion, the post-remission strategy of AHCT following HiDAC consolidation in CBF-AML was feasible and efficacious. Assays for MRD and KIT mutation may guide selection of patients who will benefit from AHCT in CBF-AML in first CR.
Collapse
|
140
|
Craddock C, Jackson A, Loke J, Siddique S, Hodgkinson A, Mason J, Andrew G, Nagra S, Malladi R, Peniket A, Gilleece M, Salim R, Tholouli E, Potter V, Crawley C, Wheatley K, Protheroe R, Vyas P, Hunter A, Parker A, Wilson K, Pavlu J, Byrne J, Dillon R, Khan N, McCarthy N, Freeman SD. Augmented Reduced-Intensity Regimen Does Not Improve Postallogeneic Transplant Outcomes in Acute Myeloid Leukemia. J Clin Oncol 2021; 39:768-778. [PMID: 33373276 PMCID: PMC8078252 DOI: 10.1200/jco.20.02308] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/10/2020] [Accepted: 12/04/2020] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Reduced-intensity conditioning (RIC) regimens have extended the curative potential of allogeneic stem-cell transplantation to older adults with high-risk acute myeloid leukemia (AML) and myelodysplasia (MDS) but are associated with a high risk of disease relapse. Strategies to reduce recurrence are urgently required. Registry data have demonstrated improved outcomes using a sequential transplant regimen, fludarabine/amsacrine/cytarabine-busulphan (FLAMSA-Bu), but the impact of this intensified conditioning regimen has not been studied in randomized trials. PATIENTS AND METHODS Two hundred forty-four patients (median age, 59 years) with high-risk AML (n = 164) or MDS (n = 80) were randomly assigned 1:1 to a fludarabine-based RIC regimen or FLAMSA-Bu. Pretransplant measurable residual disease (MRD) was monitored by flow cytometry (MFC-MRD) and correlated with outcome. RESULTS There was no difference in 2-year overall survival (hazard ratio 1.05 [85% CI, 0.80 to 1.38] P = .81) or cumulative incidence of relapse (CIR) (hazard ratio 0.94 [95%CI, 0.60 to 1.46] P = .81) between the control and FLAMSA-Bu arms. Detectable pretransplant MFC-MRD was associated with an increased CIR (2-year CIR 41.0% v 20.0%, P = .01) in the overall trial cohort with a comparable prognostic impact when measured by an unsupervised analysis approach. There was no evidence of interaction between MRD status and conditioning regimen intensity for relapse or survival. Acquisition of full donor T-cell chimerism at 3 months abrogated the adverse impact of pretransplant MRD on CIR and overall survival. CONCLUSION The intensified RIC conditioning regimen, FLAMSA-Bu, did not improve outcomes in adults transplanted for high-risk AML or MDS regardless of pretransplant MRD status. Our data instead support the exploration of interventions with the ability to accelerate acquisition of full donor T-cell chimerism as a tractable strategy to improve outcomes in patients allografted for AML.
Collapse
Affiliation(s)
- Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- Cancer Research UK Clinical Trials Unit, University of Birmingham, United Kingdom
| | - Aimee Jackson
- Cancer Research UK Clinical Trials Unit, University of Birmingham, United Kingdom
| | - Justin Loke
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Shamyla Siddique
- Cancer Research UK Clinical Trials Unit, University of Birmingham, United Kingdom
| | - Andrea Hodgkinson
- Cancer Research UK Clinical Trials Unit, University of Birmingham, United Kingdom
| | - John Mason
- Cancer Research UK Clinical Trials Unit, University of Birmingham, United Kingdom
| | - Georgia Andrew
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Sandeep Nagra
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Ram Malladi
- Addenbrookes Hospital, Cambridge, United Kingdom
| | | | | | | | | | | | | | - Keith Wheatley
- Cancer Research UK Clinical Trials Unit, University of Birmingham, United Kingdom
| | | | | | - Ann Hunter
- Leicester Royal Infirmary, United Kingdom
| | - Anne Parker
- Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | | | - Jiri Pavlu
- Imperial College Hospital, London, Unite Kingdom
| | - Jenny Byrne
- Centre for Clinical Haematology, Nottingham, United Kingdom
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Naeem Khan
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Nicholas McCarthy
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Sylvie D. Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| |
Collapse
|
141
|
Hilberink JR, Morsink LM, van der Velden WJ, Mulder AB, Hazenberg CL, de Groot M, Choi G, Schuringa JJ, Meijer K, Blijlevens NM, Ammatuna E, Huls G. Pretransplantation MRD in Older Patients With AML After Treatment With Decitabine or Conventional Chemotherapy. Transplant Cell Ther 2021; 27:246-252. [DOI: 10.1016/j.jtct.2020.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/25/2022]
|
142
|
Rimando JC, Christopher MJ, Rettig MP, DiPersio JF. Biology of Disease Relapse in Myeloid Disease: Implication for Strategies to Prevent and Treat Disease Relapse After Stem-Cell Transplantation. J Clin Oncol 2021; 39:386-396. [PMID: 33434062 PMCID: PMC8462627 DOI: 10.1200/jco.20.01587] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Joseph C. Rimando
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Matthew J. Christopher
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Michael P. Rettig
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
143
|
Chojecki A. Minimal Residual Disease in Acute Myelogenous Leukemia Is Predictive, Unless You Are Over Age 60 and Treated with Decitabine. Transplant Cell Ther 2021; 27:197-198. [PMID: 33781515 DOI: 10.1016/j.jtct.2021.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Affiliation(s)
- Aleksander Chojecki
- Department of Hematology and Cellular Therapy, Levine Cancer Institute Atrium Health, Charlotte, North Carolina.
| |
Collapse
|
144
|
Hansen DK, Kim J, Thompson Z, Hussaini M, Nishihori T, Ahmad A, Elmariah H, Faramand R, Mishra A, Davila ML, Khimani F, Lazaryan A, Sallman D, Liu H, Perez LE, Fernandez H, Nieder ML, Lancet JE, Pidala JA, Anasetti C, Bejanyan N. ELN 2017 Genetic Risk Stratification Predicts Survival of Acute Myeloid Leukemia Patients Receiving Allogeneic Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2021; 27:256.e1-256.e7. [PMID: 33781526 DOI: 10.1016/j.jtct.2020.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/11/2020] [Accepted: 12/19/2020] [Indexed: 12/17/2022]
Abstract
European LeukemiaNet (ELN) 2017 risk stratification by genetics is prognostic of outcomes in patients with acute myeloid leukemia (AML). However, the prognostic impact of the 2017 ELN genetic risk stratification after allogeneic hematopoietic cell transplantation (alloHCT) is not well established. We examined the effect of 2017 ELN genetic risk stratification on alloHCT outcomes of AML. We included 500 adult (≥18 years) AML patients in first (n = 370) or second (n = 130) complete remission receiving alloHCT from 2005 to 2016. Patients were classified into favorable (12%), intermediate (57%), and adverse (32%) 2017 ELN risk groups. The Cox proportional hazard model was used to conduct the multivariable analyses of leukemia-free survival (LFS) and overall survival (OS). Relapse and nonrelapse mortality were analyzed by the Fine-Gray regression model. OS at 2 years was 72% in the favorable versus 60% in the intermediate versus 45% in the adverse risk groups (P < .001). In multivariable analyses, the 2017 ELN classifier was an independent predictor of OS after alloHCT with significantly higher overall mortality in the intermediate (hazard ratio [HR] = 1.68, 95% confidence interval [CI], 1.06-2.68; P = .03) and adverse (HR = 2.50, 95% CI, 1.54-4.06; P < .001) risk groups compared to the favorable risk group. Similarly, LFS was worse in the intermediate (HR = 1.63, 95%, CI 1.06-2.53; P = .03) and adverse (HR 2.23, 95% CI, 1.41-3.54; P < .001) risk groups while relapse was higher in the adverse risk group (HR = 2.36, 95% CI, 1.28-4.35; P = .006) as compared to the favorable risk group. These data highlight the prognostic impact of the 2017 ELN genetic risk stratification on the survival of AML patients after alloHCT. Patients in the adverse risk group had the highest risk of relapse and worst survival. Thus the 2017 ELN prognostic system can help identify AML patients who may benefit from clinical trials offering relapse mitigation strategies to improve transplant outcomes.
Collapse
Affiliation(s)
- Doris K Hansen
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Zachary Thompson
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mohammad Hussaini
- Pathology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Taiga Nishihori
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anam Ahmad
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hany Elmariah
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Rawan Faramand
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Asmita Mishra
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marco L Davila
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Farhad Khimani
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aleksandr Lazaryan
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - David Sallman
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hien Liu
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Lia E Perez
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hugo Fernandez
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Michael L Nieder
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jeffrey E Lancet
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joseph A Pidala
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Claudio Anasetti
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nelli Bejanyan
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| |
Collapse
|
145
|
Singh S, Singh J, Paul D, Jain K. Treatment of Acute Leukemia During COVID-19: Focused Review of Evidence. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:289-294. [PMID: 33558204 PMCID: PMC7801183 DOI: 10.1016/j.clml.2021.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 01/28/2023]
Abstract
The Coronavirus disease-2019 (COVID-19) pandemic is an unprecedented health care crisis and has led to over 1.5 million deaths worldwide. The risk of severe COVID-19 and mortality is markedly raised in patients with cancer, prompting several collaborative groups to issue guidelines to mitigate the risk of infection by delaying or de-escalating immunosuppressive therapy. However, delayed therapy is often not feasible for patients requiring treatment for acute leukemia or stem cell transplantation. We provide a focused review of the recommendations and evidence for managing this high-risk group of patients while minimizing the risk of COVID-19 infection, and provide a small snapshot of treatment data from our center.
Collapse
Affiliation(s)
- Suvir Singh
- Department of Clinical Hematology and Stem Cell Transplantation, Dayanand Medical College and Hospital, Ludhiana, Punjab, India.
| | - Jagdeep Singh
- Department of Medical Oncology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| | - Davinder Paul
- Department of Medical Oncology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| | - Kunal Jain
- Department of Medical Oncology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| |
Collapse
|
146
|
How I treat measurable (minimal) residual disease in acute leukemia after allogeneic hematopoietic cell transplantation. Blood 2020; 135:1639-1649. [PMID: 31961921 DOI: 10.1182/blood.2019003566] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/18/2020] [Indexed: 12/12/2022] Open
Abstract
Although allogeneic hematopoietic cell transplantation (allo-HCT) is currently the standard curative treatment of acute leukemia, relapse remains unacceptably high. Measurable (minimal) residual disease (MRD) after allo-HCT may be used as a predictor of impending relapse and should be part of routine follow-up for transplanted patients. Patients with MRD may respond to therapies aiming to unleash or enhance the graft-versus-leukemia effect. However, evidence-based recommendations on how to best implement MRD testing and MRD-directed therapy after allo-HCT are lacking. Here, I describe our institutional approach to MRD monitoring for preemptive MRD-triggered intervention, using patient scenarios to illustrate the discussion.
Collapse
|
147
|
Roussel X, Daguindau E, Berceanu A, Desbrosses Y, Warda W, Neto da Rocha M, Trad R, Deconinck E, Deschamps M, Ferrand C. Acute Myeloid Leukemia: From Biology to Clinical Practices Through Development and Pre-Clinical Therapeutics. Front Oncol 2020; 10:599933. [PMID: 33363031 PMCID: PMC7757414 DOI: 10.3389/fonc.2020.599933] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Recent studies have provided several insights into acute myeloid leukemia. Studies based on molecular biology have identified eight functional mutations involved in leukemogenesis, including driver and passenger mutations. Insight into Leukemia stem cells (LSCs) and assessment of cell surface markers have enabled characterization of LSCs from hematopoietic stem and progenitor cells. Clonal evolution has been described as having an effect similar to that of microenvironment alterations. Such biological findings have enabled the development of new targeted drugs, including drug inhibitors and monoclonal antibodies with blockage functions. Some recently approved targeted drugs have resulted in new therapeutic strategies that enhance standard intensive chemotherapy regimens as well as supportive care regimens. Besides the progress made in adoptive immunotherapy, since allogenic hematopoietic stem cell transplantation enabled the development of new T-cell transfer therapies, such as chimeric antigen receptor T-cell and transgenic TCR T-cell engineering, new promising strategies that are investigated.
Collapse
Affiliation(s)
- Xavier Roussel
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Etienne Daguindau
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Ana Berceanu
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Yohan Desbrosses
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Walid Warda
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | | | - Rim Trad
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | - Eric Deconinck
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Marina Deschamps
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | - Christophe Ferrand
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
148
|
Short NJ, Zhou S, Fu C, Berry DA, Walter RB, Freeman SD, Hourigan CS, Huang X, Nogueras Gonzalez G, Hwang H, Qi X, Kantarjian H, Ravandi F. Association of Measurable Residual Disease With Survival Outcomes in Patients With Acute Myeloid Leukemia: A Systematic Review and Meta-analysis. JAMA Oncol 2020; 6:1890-1899. [PMID: 33030517 PMCID: PMC7545346 DOI: 10.1001/jamaoncol.2020.4600] [Citation(s) in RCA: 241] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022]
Abstract
IMPORTANCE Measurable residual disease (MRD) refers to neoplastic cells that cannot be detected by standard cytomorphologic analysis. In patients with acute myeloid leukemia (AML), determining the association of MRD with survival may improve prognostication and inform selection of efficient clinical trial end points. OBJECTIVE To examine the association between MRD status and disease-free survival (DFS) and overall survival (OS) in patients with AML using scientific literature. DATA SOURCES Clinical studies on AML published between January 1, 2000, and October 1, 2018, were identified via searches of PubMed, Embase, and MEDLINE. STUDY SELECTION Literature search and study screening were performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. Studies that assessed DFS or OS by MRD status in patients with AML were included. Reviews, non-English-language articles, and studies reporting only outcomes after hematopoietic cell transplantation or those with insufficient description of MRD information were excluded. DATA EXTRACTION AND SYNTHESIS Study sample size, median patient age, median follow-up time, MRD detection method, MRD assessment time points, AML subtype, specimen source, and survival outcomes were extracted. Meta-analyses were performed separately for DFS and OS using bayesian hierarchical modeling. MAIN OUTCOMES AND MEASURES Meta-analyses of survival probabilities and hazard ratios (HRs) were conducted for OS and DFS according to MRD status. RESULTS Eighty-one publications reporting on 11 151 patients were included. The average HR for achieving MRD negativity was 0.36 (95% bayesian credible interval [CrI], 0.33-0.39) for OS and 0.37 (95% CrI, 0.34-0.40) for DFS. The estimated 5-year DFS was 64% for patients without MRD and 25% for those with MRD, and the estimated OS was 68% for patients without MRD and 34% for those with MRD. The association of MRD negativity with DFS and OS was significant for all subgroups, with the exception of MRD assessed by cytogenetics or fluorescent in situ hybridization. CONCLUSIONS AND RELEVANCE The findings of this meta-analysis suggest that achievement of MRD negativity is associated with superior DFS and OS in patients with AML. The value of MRD negativity appears to be consistent across age groups, AML subtypes, time of MRD assessment, specimen source, and MRD detection methods. These results support MRD status as an end point that may allow for accelerated evaluation of novel therapies in AML.
Collapse
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Shouhao Zhou
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Chenqi Fu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Donald A. Berry
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sylvie D. Freeman
- Institute of Infection and Immunity, University of Birmingham, Birmingham, United Kingdom
| | - Christopher S. Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | | | - Hyunsoo Hwang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Xinyue Qi
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
149
|
Gaut D, Mead M. Measurable residual disease in hematopoietic stem cell transplantation-eligible patients with acute myeloid leukemia: clinical significance and promising therapeutic strategies. Leuk Lymphoma 2020; 62:8-31. [DOI: 10.1080/10428194.2020.1827251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Daria Gaut
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Monica Mead
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
150
|
Estey EH. Acute myeloid leukemia: 2021 update on risk-stratification and management. Am J Hematol 2020; 95:1368-1398. [PMID: 32833263 DOI: 10.1002/ajh.25975] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022]
Abstract
Management of AML involves choosing between purely palliative care, standard therapy and investigational therapy ("clinical trial"). Even most older patients likely benefit from treatment. Based on randomized trials CPX 351, midostaurin, gemtuzumab ozogamicin, and venetoclax, the latter three when combined with other drugs, should now be considered standard therapy. Knowledge of the likely results with these therapies is essential in deciding whether to recommend them or participate in a clinical trial, possibly including these drugs. Hence here, in the context of established prognostic algorithms, we review results with the recently- approved drugs compared with their predecessors and describe other potential options. We discuss benefit/risk ratios underlying the decision to offer allogeneic transplant and emphasize the importance of measurable residual disease. When first seeing a newly-diagnosed patient physicians must decide whether to offer conventional treatment or investigational therapy, the latter preferably in the context of a clinical trial. As noted below, such trials have led to changes in what today is considered "conventional" therapy compared to even 1-2 years ago. In older patients decision making has often included inquiring whether specific anti-AML therapy should be offered at all, rather than focusing on a purely palliative approach emphasizing transfusion and antibiotic support, with involvement of a palliative care specialist.
Collapse
Affiliation(s)
- Elihu H. Estey
- Division of Hematology University of Washington Seattle Washington
- Clinical Research Division Fred Hutchinson Cancer Research Center Seattle Washington
| |
Collapse
|