1
|
Pyzer AR, Dillon LW, Sharon E, Karrison TG, Zha Y, Fulton N, Gui G, Andrew G, Streicher H, Sweet K, Yaghmour G, Liu JJ, Jonas BA, Schimmer AD, Grant S, Zeidan AM, Hildebrandt GC, Lowrey CH, Mattison RJ, Palmisiano N, Salhotra A, Tzachanis D, Baer MR, Lin TL, Patel P, Chen H, Stadler WM, Odenike O, Larson RA, Gajewski TF, Hourigan CS, Stock W, Liu H. Randomized phase 2 study to assess the role of single-agent nivolumab to maintain remission in acute myeloid leukemia. Blood Adv 2025; 9:2144-2152. [PMID: 39928953 DOI: 10.1182/bloodadvances.2024015176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 02/12/2025] Open
Abstract
ABSTRACT We conducted a multicenter, open-label, randomized phase 2 study to assess the efficacy of nivolumab (Nivo) as maintenance therapy for patients with acute myeloid leukemia (AML) in first complete remission (CR) or CR with incomplete hematologic recovery who were not candidates for stem cell transplant. Patients were stratified and randomized to observation (Obs) or Nivo (3 mg/kg IV every 2 weeks for 46 doses). The primary end point was progression-free survival (PFS) defined as time to disease relapse or death due to any reason. Secondary end points included overall survival (OS), and evaluation of adverse events (AEs) after Nivo administration. Eighty patients were enrolled with median duration of follow-up of 24 months (33 months among survivors). PFS was 13.2 months in the Nivo arm and 10.9 months in the Obs arm. Overall PFS curves were not statistically significantly different. The median OS was 53.9 months in the Nivo arm and 30.9 months in the Obs arm. There were more AEs of any type (regardless of attribution) on the Nivo arm; 27 patients (71%) on the Nivo arm had a grade ≥3 AE compared with 5 patients (12%) on the Obs arm (P < .001). Nivo maintenance after AML chemotherapy failed to improve the PFS and OS in this randomized phase 2 study. There were increased AEs and serious AEs (SAEs) with Nivo, but these AEs and SAEs were expected and manageable. This trial was registered at www.ClinicalTrials.gov as #NCT02275533.
Collapse
Affiliation(s)
- Athalia R Pyzer
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Laura W Dillon
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Elad Sharon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Theodore G Karrison
- Department of Public Health Sciences, The University of Chicago, Chicago, IL
| | - Yuanyuan Zha
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Noreen Fulton
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Gege Gui
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Department of Biostatistics, Fralin Biomedical Research Institute, Virginia Tech Fralin Biomedical Research Institute Cancer Research Center, Washington, DC
| | - Georgia Andrew
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Howard Streicher
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kendra Sweet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - George Yaghmour
- Jane Anne Nohl Division of Hematology and Center for the Study of Blood Diseases, University of Southern California Norris Comprehensive Cancer Center of Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jane Jijun Liu
- Illinois Cancer Care and Heartland National Community of Oncology Research Programs, Peoria IL
| | - Brian A Jonas
- Division of Malignant Hematology/Cellular Therapy and Transplantation, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA
| | - Aaron D Schimmer
- Department of Medical Biophysics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Steven Grant
- Division of Hematology/Oncology and Palliative Care Virginia Commonwealth University Health Sciences Center, Richmond, VA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Gerhard C Hildebrandt
- Division of Hematology and Medical Oncology, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Christopher H Lowrey
- Division of Hematology, Dartmouth-Hitchcock Medical Center and Norris Cotton Cancer Center, Lebanon, NH
| | - Ryan J Mattison
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Neil Palmisiano
- Program for Hematologic Malignancies, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ
| | - Amandeep Salhotra
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA
| | - Dimitrios Tzachanis
- Department of Medicine, Moores Cancer Center, University of California San Diego, San Diego, CA
| | - Maria R Baer
- Department of Medicine, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| | - Tara L Lin
- Division of Hematologic Malignancies and Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | | | - Helen Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | | | | | | | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Department of Biostatistics, Fralin Biomedical Research Institute, Virginia Tech Fralin Biomedical Research Institute Cancer Research Center, Washington, DC
| | - Wendy Stock
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Hongtao Liu
- Department of Medicine, The University of Chicago, Chicago, IL
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
2
|
Chow RD, Velu P, Deihimi S, Belman J, Youn A, Shah N, Luger SM, Carroll MP, Morrissette J, Bowman RL. Persistent postremission clonal hematopoiesis shapes the relapse trajectories of acute myeloid leukemia. Blood Adv 2025; 9:1888-1899. [PMID: 39938015 PMCID: PMC12008691 DOI: 10.1182/bloodadvances.2024015149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
ABSTRACT Mutations found in acute myeloid leukemia (AML) such as DNMT3A, TET2, and ASXL1 can be found in the peripheral blood of healthy adults, a phenomenon termed clonal hematopoiesis (CH). These mutations are thought to represent the earliest genetic events in the evolution of AML. Genomic studies on samples acquired at diagnosis, remission, and at relapse have demonstrated significant stability of CH mutations after induction chemotherapy. Meanwhile, later mutations in genes such as NPM1 and FLT3 have been shown to contract at remission, and in the case of FLT3 often are absent at relapse. We sought to understand how early CH mutations influence subsequent evolutionary trajectories throughout remission and relapse in response to induction chemotherapy. We assembled a retrospective cohort of patients diagnosed with de novo AML at our institution that underwent genomic sequencing at diagnosis, remission, and/or relapse (total N = 182 patients). FLT3 and NPM1 mutations were generally eliminated at complete remission but subsequently reemerged upon relapse, whereas DNMT3A, TET2, and ASXL1 mutations often persisted through remission. CH-related mutations exhibited distinct constellations of co-occurring genetic alterations, with NPM1 and FLT3 mutations enriched in DNMT3Amut AML, whereas CBL and SRSF2 mutations were enriched in TET2mut and ASXL1mut AML, respectively. In the case of NPM1 and FLT3 mutations, these differences vanished at the time of complete remission yet readily reemerged upon relapse, indicating the reproducible nature of these genetic interactions. Thus, CH-associated mutations that likely precede malignant transformation subsequently shape the evolutionary trajectories of AML through diagnosis, therapy, and relapse.
Collapse
Affiliation(s)
- Ryan D. Chow
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Priya Velu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Weill Cornell School of Medicine, Cornell University, New York, NY
| | - Safoora Deihimi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jonathan Belman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Angela Youn
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nisargbhai Shah
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Selina M. Luger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jennifer Morrissette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Robert L. Bowman
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
3
|
Butler JT, Yashar WM, Swords R. Breaking the Bone Marrow Barrier: Peripheral Blood as a Gateway to Measurable Residual Disease Detection in Acute Myelogenous Leukemia. Am J Hematol 2025; 100:638-651. [PMID: 39777414 PMCID: PMC11886496 DOI: 10.1002/ajh.27586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025]
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous disease with high rates of relapse after initial treatment. Identifying measurable residual disease (MRD) following initial therapy is essential to assess response, predict patient outcomes, and identify those in need of additional intervention. Currently, MRD analysis relies on invasive, serial bone marrow (BM) biopsies, which complicate sample availability and processing time and negatively impact patient experience. Additionally, finding a positive result can generate more questions than answers, causing anxiety for both the patient and the provider. Peripheral blood (PB) evaluation has shown promise in detecting MRD and is now recommended by the European Leukemia Net for AML for certain genetic abnormalities. PB-based sampling allows for more frequent testing intervals and better temporal resolution of malignant expansion while sparing patients additional invasive procedures. In this review, we will discuss the current state of PB testing for MRD evaluation with a focus on next-generation sequencing methodologies that are capable of MRD detection across AML subtypes.
Collapse
Affiliation(s)
- John T. Butler
- Radiation Medicine and Applied Science, Moores Cancer CenterUniversity of California San DiegoLa JollaCaliforniaUSA
| | - William M. Yashar
- Knight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
- Division of Oncologic Sciences, Department of MedicineOregon Health & Science UniversityPortlandOregonUSA
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
| | - Ronan Swords
- Division of Oncologic Sciences, Department of MedicineOregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
4
|
Short NJ, Dillon R. Measurable residual disease monitoring in AML: Prospects for therapeutic decision-making and new drug development. Am J Hematol 2025; 100 Suppl 2:5-15. [PMID: 39319951 PMCID: PMC11832336 DOI: 10.1002/ajh.27482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/02/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Measurable residual disease (MRD) is strongly associated with risk of relapse and long-term survival outcomes in patients with acute myeloid leukemia (AML). Apart from its clear prognostic impact, MRD information is also increasingly used to guide therapeutic decision-making, including selection of appropriate patients for stem cell transplant, use of post-transplant maintenance, and candidacy for non-transplant maintenance therapies or MRD-directed clinical trials. While much progress has been made in accurately assessing MRD and understanding its clinical importance, many questions remain about how to optimize MRD testing and guide treatment decisions for individual patients. In this review, we discuss the common methods to assess MRD in AML and the prognostic impact of MRD across common clinical scenarios. We also review emerging and investigational strategies to target MRD and discuss some of the important unanswered questions and challenges in the field.
Collapse
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard Dillon
- Department of Haematology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King’s College London, London, UK
| |
Collapse
|
5
|
Gang M, Othus M, Walter RB. Significance of Measurable Residual Disease in Patients Undergoing Allogeneic Hematopoietic Cell Transplantation for Acute Myeloid Leukemia. Cells 2025; 14:290. [PMID: 39996762 PMCID: PMC11853423 DOI: 10.3390/cells14040290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) remains an important curative-intent treatment for many patients with acute myeloid leukemia (AML), but AML recurrence after allografting is common. Many factors associated with relapse after allogeneic HCT have been identified over the years. Central among these is measurable ("minimal") residual disease (MRD) as detected by multiparameter flow cytometry, quantitative polymerase chain reaction, and/or next-generation sequencing. Demonstration of a strong, independent prognostic role of pre- and early post-HCT MRD has raised hopes MRD could also serve as a predictive biomarker to inform treatment decision-making, with emerging data indicating the potential value to guide candidacy assessment for allografting as a post-remission treatment strategy, the selection of conditioning intensity, use of small molecule inhibitors as post-HCT maintenance therapy, and preemptive infusion of donor lymphocytes. Monitoring for leukemia recurrence after HCT and surrogacy for treatment response are other considerations for the clinical use of MRD data. In this review, we will outline the current landscape of MRD as a biomarker for patients with AML undergoing HCT and discuss areas of uncertainty and ongoing research.
Collapse
Affiliation(s)
- Margery Gang
- Hematology and Oncology Fellowship Program, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA 98109, USA;
| | - Megan Othus
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA;
| | - Roland B. Walter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
6
|
Roshal M, Gao Q. Flow cytometry evaluation of acute myeloid leukemia minimal residual disease based on an understanding of the normal maturation patterns in the blast compartments. Am J Clin Pathol 2025:aqae187. [PMID: 39921543 DOI: 10.1093/ajcp/aqae187] [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: 11/09/2024] [Accepted: 01/03/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVE Detection of minimal/measurable disease (MRD) in acute myeloid leukemia (AML) is critical for both clinical decision-making and prognostication, yet remains a challenge. Flow cytometry is a well-established method for MRD detection. Flow cytometric (FC) evaluation of MRD must consider a complex maturational pattern of normal hematopoietic development to separate normal from abnormal progenitors. Here, we offer an example of an interpretive approach based on a thorough understanding of stage- and lineage-specific hematopoietic maturation. METHODS We provide a comprehensive overview of blast maturation from early precursors (hematopoietic stem cells) to committed late-stage unilineage progenitors and commonly observed stage-specific abnormalities based on cases we have encountered in practice. We emphasize the importance of stage-specific comparisons for accurate MRD detection by flow cytometry. RESULTS The AML blasts almost invariably show abnormal phenotypes, and the phenotypes may evolve upon therapy. The detected phenotypes are necessarily confined to the target antigens included in the panel. It is therefore critical to evaluate a range of antigens to establish a specific stage/state of lineage commitment and detect potential common abnormalities. Moreover, enough cells must be acquired to allow for the detection of MRD at desired levels. Significant technical and analytical validation is critical. CONCLUSIONS Flow cytometry offers a powerful single-cell-based platform for MRD detection in AML, and the results have been proven critical for disease management. Leukemia-associated phenotype-informed difference from the normal approach presented in this review presents an analytical framework for sensitive and accurate MRD detection.
Collapse
Affiliation(s)
- Mikhail Roshal
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, US
| | - Qi Gao
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, US
| |
Collapse
|
7
|
Gui G, Ravindra N, Hegde PS, Andrew G, Mukherjee D, Wong Z, Auletta JJ, El Chaer F, Chen EC, Chen YB, Corner A, Devine SM, Iyer SG, Jimenez Jimenez AM, De Lima MJG, Litzow MR, Kebriaei P, Saber W, Spellman SR, Zeger SL, Page KM, Dillon LW, Hourigan CS. Measurable residual mutated IDH2 before allogeneic transplant for acute myeloid leukemia. Bone Marrow Transplant 2025; 60:144-153. [PMID: 39455897 PMCID: PMC11810785 DOI: 10.1038/s41409-024-02449-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Routine genetic profiling of acute myeloid leukemia (AML) at initial diagnosis has allowed subgroup specific prognostication, drug development, and clinical management strategies. The optimal approach for treatment response assessment for AML subgroups has not yet however been determined. A nationwide cohort of 257 adult patients in first remission (CR1) from AML associated with an IDH2 mutation (IDH2m) undergoing allogeneic transplant during the period 2013-2019 in the United States had rates of relapse and survival three years after transplantation of 24% and 71%, respectively. Pre-transplant clinical flow cytometry assessment was not useful in stratifying patients based on risk of post-transplant relapse or death. DNA-sequencing was performed on CR1 blood collected within 100 days before transplant. Persistent detection of IDH2m was common (51%) and associated with increased relapse and death compared to testing negative. Co-mutation at initial diagnosis with mutated NPM1 and/or FLT3-ITD was common in this cohort (41%) and use of these validated MRD markers provided superior stratification compared to IDH2m testing. Patients testing negative for IDH2m prior to transplant had low relapse-related death, regardless of conditioning intensity. Post-transplant relapse rates for those with persistently detectable IDH2m in pre-transplant remission were lower after the FDA approval of enasidenib in August 2017.
Collapse
Affiliation(s)
- Gege Gui
- Fralin Biomedical Research Institute, Virginia Tech FBRI Cancer Research Center, Washington, DC, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Niveditha Ravindra
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pranay S Hegde
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Georgia Andrew
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Devdeep Mukherjee
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zoë Wong
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffery J Auletta
- Center for International Blood and Marrow Transplant Research, NMDP, Minneapolis, MN, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Evan C Chen
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yi-Bin Chen
- Massachusetts General Hospital, Boston, MA, USA
| | | | - Steven M Devine
- Center for International Blood and Marrow Transplant Research, NMDP, Minneapolis, MN, USA
| | - Sunil G Iyer
- Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | - Partow Kebriaei
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wael Saber
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stephen R Spellman
- Center for International Blood and Marrow Transplant Research, NMDP, Minneapolis, MN, USA
| | - Scott L Zeger
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kristin M Page
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Laura W Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christopher S Hourigan
- Fralin Biomedical Research Institute, Virginia Tech FBRI Cancer Research Center, Washington, DC, USA.
| |
Collapse
|
8
|
Shaffer BC, Kebriaei P, de Lima M, Jimenez Jimenez AM. Measurable residual disease testing and allogeneic hematopoietic cell transplantation for AML: adapting Pre-MEASURE to clinical practice. Bone Marrow Transplant 2025; 60:128-134. [PMID: 39562717 PMCID: PMC11810777 DOI: 10.1038/s41409-024-02481-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Measurable residual disease (MRD) testing in patients with acute myelogenous leukemia (AML) represents a heterogenous assessment process designed to quantify leukemia-specific biomarkers that are not ascertainable by routine pathologic evaluation. The most common tools used to assess MRD are multiparameter flow cytometry (MPFC), and polymerase chain reaction (PCR) based tools, including quantitative or digital droplet PCR (qPCR, ddPCR), or next-generation sequencing (NGS) technologies. Collectively, MRD assessments have become an important clinical tool in the management of patients with AML. Despite progress, significant questions remain with respect to the appropriate timing, frequency, and methodology of MRD assessment, and whether or how to adapt therapy based on MRD results. Recent data from the Pre-MEASURE study, a retrospective cohort analysis of error corrected NGS based MRD assessment prior to allogeneic hematopoietic cell transplantation (alloHCT) in patients with AML, provides additional key information with respect to the emerging role of NGS-based technology in MRD assessment. In the context of this review, we evaluate the Pre-MEASURE study as well as other recent, high-quality assessments of MRD in AML. Our focus is to provide a practical assessment of the use of emerging MRD technologies in patients with AML with an emphasis on the role of peri-transplant MRD for the practicing clinician.
Collapse
Affiliation(s)
- Brian C Shaffer
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marcos de Lima
- Division of Hematology and Oncology, and Stem Cell Transplant Program, Case Western Reserve University, Cleveland, OH, USA
| | - Antonio M Jimenez Jimenez
- Division of Transplantation and Cell Therapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
9
|
Blackmon A, Afkhami M, Yang D, Mokhtari S, Samara Y, Pourhassan H, Ball B, Salhotra A, Agrawal V, Sandhu K, Desai A, Otoukesh S, Arslan S, Amanam I, Koller P, Tinajero J, Aribi A, Aldoss I, Becker P, Artz A, Ali H, Stein A, Smith E, Pullarkat V, Forman SJ, Marcucci G, Nakamura R, Al Malki MM. Fludarabine melphalan reduced intensity conditioning vs radiation-based myeloablative conditioning in patients undergoing allogeneic transplantation for acute myeloid leukemia with measurable residual disease. Bone Marrow Transplant 2025; 60:165-174. [PMID: 39695333 PMCID: PMC11810767 DOI: 10.1038/s41409-024-02491-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024]
Abstract
Patients with AML and measurable residual disease (MRD) undergoing allogeneic hematopoietic cell transplantation (HCT) may benefit from myeloablative conditioning (MAC) when feasible to reduce relapse risk. Fludarabine-Melphalan (FluMel) is a common reduced intensity conditioning (RIC) regimen; however, data in MRD+ patients is sparse. We performed a retrospective review of AML patients who underwent their first HCT (2016-2021) without morphologic disease at City of Hope who had pre-transplant marrow evaluated for MRD using multicolor flow cytometry (MFC) and received radiation-based MAC or FluMel conditioning. We identified 312 patients; 44 with MRD+ disease pre-HCT. The 24-month overall survival (OS), leukemia-free survival (LFS) and cumulative incidence of relapse (CIR) were 47.7%, 40.9%, and 38.6% in MRD+, and 78.0%, 73.9%, and 14.6% in MRD- patients. Radiation-based MAC was given to 136 (43.5%) patients (n = 20 with MRD+) and FluMel was given to 174 (55.8%) patients (n = 24 with MRD+). In patients with MRD+, there was no statistically significant difference between those who received MAC vs. FluMel in 24-month OS (60% vs. 38%, p = 0.21), or CIR (35% vs. 42%, p = 0.59), respectively. Our data substantiates the adverse impact of MRD in patients with AML undergoing HCT; FluMel is a reasonable option for MRD+ patients unfit for MAC.
Collapse
Affiliation(s)
- Amanda Blackmon
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA.
| | - Michelle Afkhami
- : Department of Pathology City of Hope National Medical Center, Duarte, CA, USA
| | - Dongyun Yang
- : Department of Computational and Quantitative Medicine, Beckman Research, Institute of City of Hope, Duarte, CA, USA
| | - Sally Mokhtari
- : Department of Clinical and Translational Project Development, City of Hope National Medical Center, Duarte, CA, USA
| | - Yazeed Samara
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Hoda Pourhassan
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Brian Ball
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Amandeep Salhotra
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Vaibhav Agrawal
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Karamjeet Sandhu
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Amrita Desai
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Salman Otoukesh
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Shukaib Arslan
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Idoroenyi Amanam
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Paul Koller
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Jose Tinajero
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Ahmed Aribi
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Ibrahim Aldoss
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Pamela Becker
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Andy Artz
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Haris Ali
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Anthony Stein
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Eileen Smith
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Vinod Pullarkat
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Stephen J Forman
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Guido Marcucci
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Ryotaro Nakamura
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Monzr M Al Malki
- : Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
10
|
Radich J. Mutations and MRD: clinical implications of clonal ontogeny. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:150-157. [PMID: 39644022 DOI: 10.1182/hematology.2024000541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Measurable residual disease (MRD) is a strong but imprecise predictor of relapse in acute myeloid leukemia. Many patients fall into the outlier categories of MRD positivity without relapse or MRD negativity with relapse. Why? We will discuss these states in the context of "clonal ontogeny" examining how mutations, clonal structure, and Darwinian rules impact response, resistance, and relapse.
Collapse
Affiliation(s)
- Jerald Radich
- Translational Science & Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
11
|
Shen Q, Gong X, Feng Y, Hu Y, Wang T, Yan W, Zhang W, Qi S, Gale RP, Chen J. Measurable residual disease (MRD)-testing in haematological cancers: A giant leap forward or sideways? Blood Rev 2024; 68:101226. [PMID: 39164126 DOI: 10.1016/j.blre.2024.101226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024]
Abstract
Measurable residual disease (MRD)-testing is used in many haematological cancers to estimate relapse risk and to direct therapy. Sometimes MRD-test results are used for regulatory approval. However, some people including regulators wrongfully believe results of MRD-testing are highly accurate and of proven efficacy in directing therapy. We review MRD-testing technologies and evaluate the accuracy of MRD-testing for predicting relapse and the strength of evidence supporting efficacy of MRD-guided therapy. We show that at the individual level MRD-test results are often an inaccurate relapse predictor. Also, no convincing data indicate that increasing therapy-intensity based on a positive MRD-test reduces relapse risk or improves survival. We caution against adjusting therapy-intensity based solely on results of MRD-testing.
Collapse
Affiliation(s)
- Qiujin Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Xiaowen Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Yahui Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Tiantian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Wen Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Wei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Saibing Qi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK.
| | - Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| |
Collapse
|
12
|
Oshima MU, Higgins J, Jenkins I, Randolph T, Smith T, Valentine C, Salk J, Yeung C, Beppu L, Campbell J, Carpenter PA, Lee SJ, Flowers ME, Radich JP, Storb R. Characterization of clonal dynamics using duplex sequencing in donor-recipient pairs decades after hematopoietic cell transplantation. Sci Transl Med 2024; 16:eado5108. [PMID: 39441907 PMCID: PMC11813252 DOI: 10.1126/scitranslmed.ado5108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024]
Abstract
After allogeneic hematopoietic cell transplantation (HCT), a very small number of donor stem cells reconstitute the recipient hematopoietic system, whereas the donor is left with a near-normal pool of stem cells. We hypothesized that the increased replicative stress on transplanted donor cells in the recipient could lead to the disproportionate proliferation of clonal hematopoiesis (CH) variants. We obtained blood samples from 16 related donor-recipient pairs at a median of 33.8 years (range: 6.6 to 45.7) after HCT, including the longest surviving HCT recipients in the world. For 11 of 16 pairs, a donor sample from the time of HCT was available for comparison. We performed ultrasensitive duplex sequencing of genes recurrently mutated in myeloid malignancies and CH, as well as a set of functionally neutral genomic regions representative of human genomic content at large. CH variants were observed in all donors, even those as young as 12 years old. Where donor pre-HCT sample was available, the average mutation rate in donors compared to recipients post-HCT was similar (2.0% versus 2.6% per year, respectively) within genes recurrently mutated in myeloid malignancies. Twenty-two (5.6%) of the 393 variants shared between paired donors and recipients post-HCT showed ≥10-fold higher variant allele frequency (VAF) in the recipient. A longer time since HCT was positively associated with the expansion of shared variant VAFs in the recipient. In conclusion, even decades after HCT, there does not appear to be widespread accelerated clonal expansion in the transplanted cells, highlighting the immense regenerative capacity of the human hematopoietic system.
Collapse
Affiliation(s)
- Masumi Ueda Oshima
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195
| | | | - Isaac Jenkins
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
| | - Timothy Randolph
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
| | | | | | - Jesse Salk
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195
- TwinStrand Biosciences, Seattle, WA, 98121
| | - Cecilia Yeung
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
| | - Lan Beppu
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
| | - Judy Campbell
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
| | - Paul A. Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195
| | - Stephanie J. Lee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195
| | - Mary E. Flowers
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195
| | - Jerald P. Radich
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
| | - Rainer Storb
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98195
| |
Collapse
|
13
|
Gale RP, Phillips GL, Lazarus HM. A modest proposal to the transplant publik to prevent harm to people with acute myeloid leukaemia in 1st complete remission cured by chemotherapy. Leukemia 2024; 38:1663-1666. [PMID: 38459165 PMCID: PMC11286528 DOI: 10.1038/s41375-024-02214-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/10/2024]
Affiliation(s)
- R P Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, SW7 2AS, UK.
| | - G L Phillips
- Wake Forest School of Medicine (Emeritus), Winston-Salem, NC, USA
| | - H M Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
14
|
Radich J. Transplant, MRD, and predicting relapse in AML. Blood 2024; 144:245-247. [PMID: 39023869 DOI: 10.1182/blood.2024024870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
|
15
|
Winters AC, Minhajuddin M, Stevens BM, Major A, Bosma G, Abbott D, Miltgen N, Yuan J, Treece AL, Siegele BJ, Ewalt MD, Gutman JA, Jordan CT, Pollyea DA. Multi-gene measurable residual disease assessed by digital polymerase chain reaction has clinical and biological utility in acute myeloid leukemia patients receiving venetoclax/azacitidine. Haematologica 2024; 109:1766-1778. [PMID: 38105738 PMCID: PMC11141685 DOI: 10.3324/haematol.2023.283790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023] Open
Abstract
Venetoclax with azacitidine (ven/aza) is a lower-intensity therapeutic regimen that has been shown to improve outcomes in elderly patients with acute myeloid leukemia (AML). Measurable residual disease (MRD) using flow cytometry is a valuable tool for the prediction of relapse in AML using conventional therapies and ven/aza; however, the prognostic value for broadscale molecular MRD after ven/aza treatment is less clear. We aimed to determine the utility of retrospective assessment using multi-gene molecular MRD by droplet digital polymerase chain reaction (ddPCR). We found this approach correlates with outcomes in a cohort of patients receiving frontline ven/aza for AML. The predictive value of ddPCR MRD persisted when NPM1 mutations were removed from analysis, as well as after adjustment for the impact of stem cell transplant on outcomes. Late achievement of MRD negativity, including after SCT, was still associated with superior outcomes compared to persistently detectable MRD. We further explored the impact of ven/aza on the burden of different classes of mutations, and identified the persistence of splicing factor mutations, commonly associated with MDS, as a consistent finding after ven/aza treatment. These data add to our understanding of the effects of ven/aza on AML disease biology and provide details on molecular depth of remission that can guide prospective trials in the future.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/diagnosis
- Neoplasm, Residual/diagnosis
- Nucleophosmin
- Sulfonamides/therapeutic use
- Sulfonamides/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Aged
- Male
- Female
- Azacitidine/therapeutic use
- Azacitidine/administration & dosage
- Middle Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Mutation
- Polymerase Chain Reaction/methods
- Prognosis
- Aged, 80 and over
- Retrospective Studies
- Adult
- Treatment Outcome
Collapse
Affiliation(s)
- Amanda C Winters
- Center for Cancer and Blood Disorders, Department of Pediatrics, University of Colorado, Aurora.
| | - Mohd Minhajuddin
- Division of Hematology, Department of Medicine, University of Colorado, Aurora
| | - Brett M Stevens
- Division of Hematology, Department of Medicine, University of Colorado, Aurora
| | - Ajay Major
- Division of Hematology, Department of Medicine, University of Colorado, Aurora
| | - Grace Bosma
- Department of Biostatistics and Informatics, University of Colorado, Aurora
| | - Diana Abbott
- Department of Biostatistics and Informatics, University of Colorado, Aurora
| | | | - Ji Yuan
- Molecular Diagnostics, Children's Hospital Colorado, Aurora
| | - Amy L Treece
- Pediatric Pathology, Children's of Alabama, Birmingham
| | | | - Mark D Ewalt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - Jonathan A Gutman
- Division of Hematology, Department of Medicine, University of Colorado, Aurora
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado, Aurora
| | - Daniel A Pollyea
- Division of Hematology, Department of Medicine, University of Colorado, Aurora
| |
Collapse
|
16
|
Chen J, Gale RP, Hu Y, Yan W, Wang T, Zhang W. Measurable residual disease (MRD)-testing in haematological and solid cancers. Leukemia 2024; 38:1202-1212. [PMID: 38637690 PMCID: PMC11147778 DOI: 10.1038/s41375-024-02252-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Affiliation(s)
- Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wen Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tiantian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
17
|
Law AD, Mattsson JI. Transplant without salvage: cut out the middleman. Lancet Haematol 2024; 11:e310-e311. [PMID: 38583456 DOI: 10.1016/s2352-3026(24)00072-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024]
Affiliation(s)
- Arjun Datt Law
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Maragaret Cancer Centre, Toronto, ON M5G 2M9, Canada; Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jonas Ingemar Mattsson
- Hans Messner Allogeneic Blood and Marrow Transplant Program, Princess Maragaret Cancer Centre, Toronto, ON M5G 2M9, Canada; Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Toronto, ON, Canada.
| |
Collapse
|
18
|
Zhang L, Deeb G, Deeb KK, Vale C, Peker Barclift D, Papadantonakis N. Measurable (Minimal) Residual Disease in Myelodysplastic Neoplasms (MDS): Current State and Perspectives. Cancers (Basel) 2024; 16:1503. [PMID: 38672585 PMCID: PMC11048433 DOI: 10.3390/cancers16081503] [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: 02/17/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Myelodysplastic Neoplasms (MDS) have been traditionally studied through the assessment of blood counts, cytogenetics, and morphology. In recent years, the introduction of molecular assays has improved our ability to diagnose MDS. The role of Measurable (minimal) Residual Disease (MRD) in MDS is evolving, and molecular and flow cytometry techniques have been used in several studies. In this review, we will highlight the evolving concept of MRD in MDS, outline the various techniques utilized, and provide an overview of the studies reporting MRD and the correlation with outcomes.
Collapse
Affiliation(s)
- Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - George Deeb
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kristin K. Deeb
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Colin Vale
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Deniz Peker Barclift
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nikolaos Papadantonakis
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
19
|
Walter RB. Perspective on measurable residual disease testing in acute myeloid leukemia. Leukemia 2024; 38:10-13. [PMID: 37973819 DOI: 10.1038/s41375-023-02084-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Affiliation(s)
- Roland B Walter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
20
|
Li Y, Dou L, Nervi C. Editorial: Genetic/epigenetic mechanisms and related clinical strategy in leukemia. Front Oncol 2023; 13:1275992. [PMID: 37671059 PMCID: PMC10476082 DOI: 10.3389/fonc.2023.1275992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 09/07/2023] Open
Affiliation(s)
- Yonghui Li
- Central Laboratory, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Liping Dou
- Department of Hematology, People’s Liberation Army General Hospital, Beijing, China
| | - Clara Nervi
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| |
Collapse
|