1
|
Tedjaseputra A, Russell N, Dillon R. SOHO State of the Art Updates and Next Questions: Pre-emptive Therapy at Molecular Measurable Residual Disease Failure in Acute Myeloid Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:737-751. [PMID: 38734498 DOI: 10.1016/j.clml.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 05/13/2024]
Abstract
Molecular measurable residual disease (MRD, eg, by real-time quantitative polymerase chain reaction, RT-qPCR), is an integral part of response assessment in acute myeloid leukemia (AML) with established prognostic and evolving therapeutic significance. MRD failure can occur through several pathways (namely MRD persistence at the end of treatment at a high level, MRD progression from a low level or MRD re-emergence during follow up; the latter two constitute MRD relapse as defined by the European Leukemia Net) and is clinically actionable, with survival benefit reported in AML subgroups. Selection of pre-emptive therapy at MRD failure relies upon an integrated clinico-molecular assessment and is subset-specific. In acute promyelocytic leukemia, arsenic trioxide-based regimen for MRD failure following frontline treatment with all-trans-retinoic acid plus chemotherapy represents standard of care, while hypomethylating agents (eg, azacitidine), salvage chemotherapy (eg, FLAG-IDA) and venetoclax-based regimens are effective in NPM1-mutated AML. Specific inhibitors of FLT3 have emerging use in FLT3-mutated AML and are associated with minimal toxicity. Furthermore, immunotherapeutic approaches such as donor lymphocyte infusions and interferon-⍺ are efficacious options in the post-allogeneic-HSCT settings. Enrollment into clinical trials with genomic-guided assignment of pre-emptive therapy at MRD failure should be prioritized. Finally, with the emergence of novel agents (eg, menin inhibitors) and approaches (eg, adoptive cellular and immunological therapy), an exciting future lies ahead where a broad array of highly active pre-emptive therapeutic options will likely be clinically applicable to a wide range of AML subsets.
Collapse
Affiliation(s)
- Aditya Tedjaseputra
- 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; Monash Haematology, Melbourne, Australia
| | - Nigel Russell
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - 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
|
2
|
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 2024:10.1038/s41409-024-02449-2. [PMID: 39455897 DOI: 10.1038/s41409-024-02449-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [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
|
3
|
Hegde PS, Andrew G, Gui G, Ravindra N, Mukherjee D, Wong ZC, Auletta JJ, El Chaer F, Corner A, Devine SM, Jimenez Jimenez AM, De Lima MJG, Litzow MR, Kebriaei P, Saber W, Spellman SR, Zeger SL, Page KM, Dillon LW, Hourigan CS. Measurable residual FLT3 tyrosine kinase domain mutations before allogeneic transplant for acute myeloid leukemia. Bone Marrow Transplant 2024:10.1038/s41409-024-02444-7. [PMID: 39424958 DOI: 10.1038/s41409-024-02444-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Affiliation(s)
- 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
| | - 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
| | - Devdeep Mukherjee
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zoë C 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
| | | | | | - Steven M Devine
- Center for International Blood and Marrow Transplant Research, NMDP, Minneapolis, MN, 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
|
4
|
Short NJ, Dillon R. Measurable residual disease monitoring in AML: Prospects for therapeutic decision-making and new drug development. Am J Hematol 2024. [PMID: 39319951 DOI: 10.1002/ajh.27482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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, Texas, USA
| | - 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
|
Oduro KA, Spivey T, Moore EM, Meyerson H, Yoest J, Tomlinson B, Beck R, Alouani D, Sadri N. Clonal Dynamics and Relapse Risk Revealed by High-Sensitivity FLT3-Internal Tandem Duplication Detection in Acute Myeloid Leukemia. Mod Pathol 2024; 37:100534. [PMID: 38852814 DOI: 10.1016/j.modpat.2024.100534] [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: 02/27/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 06/11/2024]
Abstract
The ability to detect low-level disease is key to our understanding of clonal heterogeneity in acute myeloid leukemia (AML) and residual disease that elude conventional assays and seed relapse. We developed a high-sensitivity next-generation sequencing (HS-NGS) clinical assay, able to reliably detect low levels (1 × 10-5) of FLT3-ITD, a frequent, therapeutically targetable and prognostically relevant mutation in AML. By applying this assay to 289 longitudinal samples from 62 patients at initial diagnosis and/or clinical follow-up (mean follow-up of 22 months), we reveal the frequent occurrence of FLT3-ITD subclones at diagnosis and demonstrate a significantly decreased relapse risk when FLT3-ITD is cleared after induction or thereafter. We perform pairwise sequencing of diagnosis and relapse samples from 23 patients to uncover more detailed patterns of FLT3-ITD clonal evolution at relapse than is detectable by less-sensitive assays. Finally, we show that rising ITD level during consecutive biopsies is a harbinger of impending relapse. Our findings corroborate the emerging clinical utility of high-sensitivity FLT3-ITD testing and expands our understanding of clonal dynamics in FLT3-ITD-positive AML.
Collapse
Affiliation(s)
- Kwadwo Asare Oduro
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Pathology and Laboratory Medicine, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio.
| | - Theresa Spivey
- Department of Pathology and Laboratory Medicine, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Erika M Moore
- Department of Pathology and Laboratory Medicine, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Howard Meyerson
- Department of Pathology and Laboratory Medicine, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Jennifer Yoest
- Department of Pathology and Laboratory Medicine, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Benjamin Tomlinson
- Department of Hematology/Oncology, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Rose Beck
- Department of Pathology and Laboratory Medicine, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - David Alouani
- Department of Pathology and Laboratory Medicine, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Navid Sadri
- Department of Pathology and Laboratory Medicine, University Hospitals Cleveland Medical Center & Case Western Reserve University School of Medicine, Cleveland, Ohio.
| |
Collapse
|
6
|
Wienecke CP, Heida B, Venturini L, Gabdoulline R, Krüger K, Teich K, Büttner K, Wichmann M, Puppe W, Neziri B, Reuter M, Dammann E, Stadler M, Ganser A, Hambach L, Thol F, Heuser M. Clonal relapse dynamics in acute myeloid leukemia following allogeneic hematopoietic cell transplantation. Blood 2024; 144:296-307. [PMID: 38669617 DOI: 10.1182/blood.2023022697] [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: 09/28/2023] [Revised: 03/06/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
ABSTRACT Patients with acute myeloid leukemia (AML) who experience relapse following allogeneic hematopoietic cell transplantation (alloHCT) face unfavorable outcomes regardless of the chosen relapse treatment. Early detection of relapse at the molecular level by measurable residual disease (MRD) assessment enables timely intervention, which may prevent hematological recurrence of the disease. It remains unclear whether molecular MRD assessment can detect MRD before impending relapse and, if so, how long in advance. This study elucidates the molecular architecture and kinetics preceding AML relapse by using error-corrected next-generation sequencing (NGS) in 74 patients with AML relapsing after alloHCT, evaluating 140 samples from peripheral blood collected 0.6 to 14 months before relapse. At least 1 MRD marker became detectable in 10%, 38%, and 64% of patients at 6, 3, and 1 month before relapse, respectively. By translating these proportions into monitoring intervals, 38% of relapses would have been detected through MRD monitoring every 3 months, whereas 64% of relapses would have been detected with monthly intervals. The relapse kinetics after alloHCT are influenced by the functional class of mutations and their stability during molecular progression. Notably, mutations in epigenetic modifier genes exhibited a higher prevalence of MRD positivity and greater stability before relapse, whereas mutations in signaling genes demonstrated a shorter lead time to relapse. Both DTA (DNMT3A, TET2, and ASXL1) and non-DTA mutations displayed similar relapse kinetics during the follow-up period after alloHCT. Our study sets a framework for MRD monitoring after alloHCT by NGS, supporting monthly monitoring from peripheral blood using all variants that are known from diagnosis.
Collapse
Affiliation(s)
- Clara Philine Wienecke
- 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
| | - Letizia Venturini
- 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
| | - Katja Krüger
- 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
| | - Konstantin Büttner
- 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
| | - Wolfram Puppe
- Department of Virology, Hannover Medical School, Hannover, Germany
| | - Blerina Neziri
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Marlene Reuter
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Elke Dammann
- 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
| | - Arnold Ganser
- 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
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
7
|
Tiong IS, Hiwase DK, Abro E, Bajel A, Palfreyman E, Beligaswatte A, Reynolds J, Anstee N, Nguyen T, Loo S, Chua CC, Ashby M, Wiltshire KM, Fleming S, Fong CY, Teh TC, Blombery P, Dillon R, Ivey A, Wei AH. Targeting Molecular Measurable Residual Disease and Low-Blast Relapse in AML With Venetoclax and Low-Dose Cytarabine: A Prospective Phase II Study (VALDAC). J Clin Oncol 2024; 42:2161-2173. [PMID: 38427924 PMCID: PMC11191043 DOI: 10.1200/jco.23.01599] [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: 07/27/2023] [Revised: 11/21/2023] [Accepted: 12/18/2023] [Indexed: 03/03/2024] Open
Abstract
PURPOSE A prospective phase II study examined the safety and efficacy of venetoclax combined with low-dose cytarabine (LDAC) in AML at first measurable residual disease (MRD) or oligoblastic relapse. METHODS Patients with either MRD (≥1 log10 rise) or oligoblastic relapse (blasts 5%-15%) received venetoclax 600 mg once daily D1-28 plus LDAC once daily D1-10 in 28-day cycles. The primary objective was MRD response in the MRD relapse cohort or complete remission (CR/CRh/CRi) in the oligoblastic relapse cohort. RESULTS Forty-eight adults with either MRD (n = 26) or oligoblastic (n = 22) relapse were enrolled. Median age was 67 years (range, 18-80) and 94% had received previous intensive chemotherapy. Patients received a median of four cycles of therapy; 17% completed ≥12 cycles. Patients with oligoblastic relapse had more grade ≥3 anemia (32% v 4%; P = .02) and infections (36% v 8%; P = .03), whereas grade 4 neutropenia (32 v 23%) or thrombocytopenia (27 v 15%) were comparable with the MRD relapse cohort. Markers of molecular MRD relapse included mutant NPM1 (77%), CBFB::MYH11 (4%), RUNX1::RUNX1T1 (4%), or KMT2A::MLLT3 (4%). Three patients with a log10 rise in IDH1/2 (12%) were included. By cycle 2 in the MRD relapse cohort, a log10 reduction in MRD was observed in 69%; 46% achieved MRD negative remission. In the oligoblastic relapse cohort, 73% achieved CR/CRh/CRi. Overall, 21 (44%) underwent hematopoietic cell transplantation. Median overall survival (OS) was not reached in either cohort. Estimated 2-year OS rate was 67% (95% CI, 50 to 89) in the MRD and 53% (95% CI, 34 to 84) in the oligoblastic relapse cohorts. CONCLUSION For AML in first remission and either MRD or oligoblastic relapse, venetoclax plus LDAC is well tolerated and highly effective.
Collapse
MESH Headings
- Humans
- Aged
- Middle Aged
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Neoplasm, Residual
- Cytarabine/administration & dosage
- Sulfonamides/administration & dosage
- Sulfonamides/adverse effects
- Adult
- Female
- Male
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Aged, 80 and over
- Prospective Studies
- Nucleophosmin
- Young Adult
- Adolescent
Collapse
Affiliation(s)
- Ing Soo Tiong
- The Alfred Hospital and Monash University, Melbourne, Australia
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
| | - Devendra K. Hiwase
- Royal Adelaide Hospital, Adelaide, Australia
- University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Emad Abro
- Princess Alexandra Hospital, Queensland, Australia
| | - Ashish Bajel
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
| | | | - Ashanka Beligaswatte
- University of Adelaide, Adelaide, Australia
- Flinders Medical Centre, Bedford Park, Australia
| | - John Reynolds
- The Alfred Hospital and Monash University, Melbourne, Australia
| | - Natasha Anstee
- The University of Melbourne, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Tamia Nguyen
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
| | - Sun Loo
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- The Northern Hospital, Melbourne, Australia
| | - Chong Chyn Chua
- The Alfred Hospital and Monash University, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- The Northern Hospital, Melbourne, Australia
| | - Michael Ashby
- The Alfred Hospital and Monash University, Melbourne, Australia
| | | | - Shaun Fleming
- The Alfred Hospital and Monash University, Melbourne, Australia
| | - Chun Y. Fong
- Austin Health and Olivia Newton John Cancer Research Institute, Melbourne, Australia
| | - Tse-Chieh Teh
- The Alfred Hospital and Monash University, Melbourne, Australia
- Box Hill Hospital, Melbourne, Australia
| | - Piers Blombery
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Guy's Hospital, London, United Kingdom
| | - Adam Ivey
- The Alfred Hospital and Monash University, Melbourne, Australia
| | - Andrew H. Wei
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| |
Collapse
|
8
|
Li EW, Tran NYK, McCulloch D, Krigstein M, Catalano A, Othman J, Abadir E, Smith C, Iland H. FLT3-TKD Measurable Residual Disease Detection Using Droplet Digital PCR and Clinical Applications in Acute Myeloid Leukemia. Int J Mol Sci 2024; 25:5771. [PMID: 38891959 PMCID: PMC11171966 DOI: 10.3390/ijms25115771] [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: 03/28/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
The tyrosine kinase domain of the FMS-Like tyrosine kinase 3 (FLT3-TKD) is recurrently mutated in acute myeloid leukemia (AML). Common molecular techniques used in its detection include PCR and capillary electrophoresis, Sanger sequencing and next-generation sequencing with recognized sensitivity limitations. This study aims to validate the use of droplet digital PCR (ddPCR) in the detection of measurable residual disease (MRD) involving the common FLT3-TKD mutations (D835Y, D835H, D835V, D835E). Twenty-two diagnostic samples, six donor controls, and a commercial D835Y positive control were tested using a commercial Bio-rad® ddPCR assay. All known variants were identified, and no false positives were detected in the wild-type control (100% specificity and sensitivity). The assays achieved a limit of detection suitable for MRD testing at 0.01% variant allelic fraction. Serial samples from seven intensively-treated patients with FLT3-TKD variants at diagnosis were tested. Five patients demonstrated clearance of FLT3-TKD clones, but two patients had FLT3-TKD persistence in the context of primary refractory disease. In conclusion, ddPCR is suitable for the detection and quantification of FLT3-TKD mutations in the MRD setting; however, the clinical significance and optimal management of MRD positivity require further exploration.
Collapse
Affiliation(s)
- Eric Wenlong Li
- Institute of Hematology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Molecular Hematology Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
| | - Ngoc Yen Kim Tran
- Molecular Hematology Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Derek McCulloch
- Institute of Hematology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Molecular Hematology Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
| | - Michael Krigstein
- Department of Hematology, St Vincent’s Hospital, Sydney, NSW 2010, Australia
| | - Alberto Catalano
- Molecular Hematology Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Jad Othman
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
- Department of Hematology, Royal North Shore Hospital, Sydney, NSW 2065, Australia
| | - Edward Abadir
- Institute of Hematology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
| | - Cheryl Smith
- Molecular Hematology Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Harry Iland
- Institute of Hematology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Molecular Hematology Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
| |
Collapse
|
9
|
Levis MJ, Hamadani M, Logan B, Jones RJ, Singh AK, Litzow M, Wingard JR, Papadopoulos EB, Perl AE, Soiffer RJ, Ustun C, Ueda Oshima M, Uy GL, Waller EK, Vasu S, Solh M, Mishra A, Muffly L, Kim HJ, Mikesch JH, Najima Y, Onozawa M, Thomson K, Nagler A, Wei AH, Marcucci G, Geller NL, Hasabou N, Delgado D, Rosales M, Hill J, Gill SC, Nuthethi R, King D, Wittsack H, Mendizabal A, Devine SM, Horowitz MM, Chen YB. Gilteritinib as Post-Transplant Maintenance for AML With Internal Tandem Duplication Mutation of FLT3. J Clin Oncol 2024; 42:1766-1775. [PMID: 38471061 PMCID: PMC11095884 DOI: 10.1200/jco.23.02474] [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: 11/15/2023] [Revised: 11/29/2023] [Accepted: 12/28/2023] [Indexed: 03/14/2024] Open
Abstract
PURPOSE Allogeneic hematopoietic cell transplantation (HCT) improves outcomes for patients with AML harboring an internal tandem duplication mutation of FLT3 (FLT3-ITD) AML. These patients are routinely treated with a FLT3 inhibitor after HCT, but there is limited evidence to support this. Accordingly, we conducted a randomized trial of post-HCT maintenance with the FLT3 inhibitor gilteritinib (ClinicalTrials.gov identifier: NCT02997202) to determine if all such patients benefit or if detection of measurable residual disease (MRD) could identify those who might benefit. METHODS Adults with FLT3-ITD AML in first remission underwent HCT and were randomly assigned to placebo or 120 mg once daily gilteritinib for 24 months after HCT. The primary end point was relapse-free survival (RFS). Secondary end points included overall survival (OS) and the effect of MRD pre- and post-HCT on RFS and OS. RESULTS Three hundred fifty-six participants were randomly assigned post-HCT to receive gilteritinib or placebo. Although RFS was higher in the gilteritinib arm, the difference was not statistically significant (hazard ratio [HR], 0.679 [95% CI, 0.459 to 1.005]; two-sided P = .0518). However, 50.5% of participants had MRD detectable pre- or post-HCT, and, in a prespecified subgroup analysis, gilteritinib was beneficial in this population (HR, 0.515 [95% CI, 0.316 to 0.838]; P = .0065). Those without detectable MRD showed no benefit (HR, 1.213 [95% CI, 0.616 to 2.387]; P = .575). CONCLUSION Although the overall improvement in RFS was not statistically significant, RFS was higher for participants with detectable FLT3-ITD MRD pre- or post-HCT who received gilteritinib treatment. To our knowledge, these data are among the first to support the effectiveness of MRD-based post-HCT therapy.
Collapse
Affiliation(s)
| | | | - Brent Logan
- CIBMTR/Medical College of Wisconsin, Milwaukee, WI
| | | | | | | | | | | | | | | | | | | | | | | | | | - Melhem Solh
- Northside Hospital Cancer Institute, Atlanta, GA
| | | | | | - Hee-Je Kim
- Catholic Hematology Hospital, Seoul St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Yuho Najima
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | | | | | - Arnon Nagler
- Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Andrew H. Wei
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital, Walter and Eliza Hill Institute of Medical Research and University of Melbourne, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Lucero J, Alhumaid M, Novitzky-Basso I, Capo-Chichi JM, Stockley T, Gupta V, Bankar A, Chan S, Schuh AC, Minden M, Mattsson J, Kumar R, Sibai H, Tierens A, Kim DDH. Flow cytometry-based measurable residual disease (MRD) analysis identifies AML patients who may benefit from allogeneic hematopoietic stem cell transplantation. Ann Hematol 2024; 103:1187-1196. [PMID: 38291275 DOI: 10.1007/s00277-024-05639-6] [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: 09/21/2023] [Accepted: 01/21/2024] [Indexed: 02/01/2024]
Abstract
Measurable residual disease (MRD) monitoring independently predicts long-term outcomes in patients with acute myeloid leukemia (AML). Of the various modalities available, multiparameter flow cytometry-based MRD analysis is widely used and relevant for patients without molecular targets. In the transplant (HCT) setting, the presence of MRD pre-HCT is associated with adverse outcomes. MRD-negative remission status pre-HCT was also associated with longer overall (OS) and progression-free survival and a lower risk of relapse. We hypothesize that the combination of disease risk and MRD at the time of first complete remission (CR1) could identify patients according to the benefit gained from HCT, especially for intermediate-risk patients. We performed a retrospective analysis comparing the outcomes of HCT versus non-HCT therapies based on MRD status in AML patients who achieved CR1. Time-dependent analysis was applied considering time-to-HCT as a time-dependent covariate and compared HCT versus non-HCT outcomes according to MRD status at CR1. Among 336 patients assessed at CR1, 35.1% were MRD positive (MRDpos) post-induction. MRDpos patients benefitted from HCT with improved OS and relapse-free survival (RFS), while no benefit was observed in MRDneg patients. In adverse-risk patients, HCT improved OS (HR for OS 0.55; p = 0.05). In intermediate-risk patients, HCT benefit was not significant for OS and RFS. Intermediate-risk MRDpos patients were found to have benefit from HCT with improved OS (HR 0.45, p = 0.04), RFS (HR 0.46, p = 0.02), and CIR (HR 0.41, p = 0.02). Our data underscore the benefit of HCT in adverse risk and MRDpos intermediate-risk AML patients.
Collapse
Affiliation(s)
- Josephine Lucero
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| | | | - Igor Novitzky-Basso
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Jose-Mario Capo-Chichi
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Tracy Stockley
- Division of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | - Vikas Gupta
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Aniket Bankar
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Steven Chan
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Andre C Schuh
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Mark Minden
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Jonas Mattsson
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Rajat Kumar
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Hassan Sibai
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Anne Tierens
- Division of Hematology and Transfusion Medicine, Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | - Dennis D H Kim
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| |
Collapse
|
11
|
Reikvam H, Dillon R. Revisiting the role of measurable residual disease in FLT3 mutated acute myelogenous leukemia. Expert Rev Hematol 2024; 17:103-106. [PMID: 38654593 DOI: 10.1080/17474086.2024.2347303] [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: 12/18/2023] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Affiliation(s)
- Håkon Reikvam
- K.G. Jebsen Center for Myeloid Blood Cancer, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
12
|
Murdock HM, Ho VT, Garcia JS. Innovations in conditioning and post-transplant maintenance in AML: genomically informed revelations on the graft-versus-leukemia effect. Front Immunol 2024; 15:1359113. [PMID: 38571944 PMCID: PMC10987864 DOI: 10.3389/fimmu.2024.1359113] [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: 12/20/2023] [Accepted: 02/20/2024] [Indexed: 04/05/2024] Open
Abstract
Acute Myeloid Leukemia (AML) is the prototype of cancer genomics as it was the first published cancer genome. Large-scale next generation/massively parallel sequencing efforts have identified recurrent alterations that inform prognosis and have guided the development of targeted therapies. Despite changes in the frontline and relapsed standard of care stemming from the success of small molecules targeting FLT3, IDH1/2, and apoptotic pathways, allogeneic stem cell transplantation (alloHSCT) and the resulting graft-versus-leukemia (GVL) effect remains the only curative path for most patients. Advances in conditioning regimens, graft-vs-host disease prophylaxis, anti-infective agents, and supportive care have made this modality feasible, reducing transplant related mortality even among patients with advanced age or medical comorbidities. As such, relapse has emerged now as the most common cause of transplant failure. Relapse may occur after alloHSCT because residual disease clones persist after transplant, and develop immune escape from GVL, or such clones may proliferate rapidly early after alloHSCT, and outpace donor immune reconstitution, leading to relapse before any GVL effect could set in. To address this issue, genomically informed therapies are increasingly being incorporated into pre-transplant conditioning, or as post-transplant maintenance or pre-emptive therapy in the setting of mixed/falling donor chimerism or persistent detectable measurable residual disease (MRD). There is an urgent need to better understand how these emerging therapies modulate the two sides of the GVHD vs. GVL coin: 1) how molecularly or immunologically targeted therapies affect engraftment, GVHD potential, and function of the donor graft and 2) how these therapies affect the immunogenicity and sensitivity of leukemic clones to the GVL effect. By maximizing the synergistic action of molecularly targeted agents, immunomodulating agents, conventional chemotherapy, and the GVL effect, there is hope for improving outcomes for patients with this often-devastating disease.
Collapse
Affiliation(s)
- H. Moses Murdock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Vincent T. Ho
- Bone Marrow Transplant Program, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Jacqueline S. Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|
13
|
Dillon LW, Higgins J, Nasif H, Othus M, Beppu L, Smith TH, Schmidt E, Valentine Iii CC, Salk JJ, Wood BL, Erba HP, Radich JP, Hourigan CS. Quantification of measurable residual disease using duplex sequencing in adults with acute myeloid leukemia. Haematologica 2024; 109:401-410. [PMID: 37534515 PMCID: PMC10828764 DOI: 10.3324/haematol.2023.283520] [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: 05/15/2023] [Accepted: 07/28/2023] [Indexed: 08/04/2023] Open
Abstract
The presence of measurable residual disease (MRD) is strongly associated with treatment outcomes in acute myeloid leukemia (AML). Despite the correlation with clinical outcomes, MRD assessment has yet to be standardized or routinely incorporated into clinical trials and discrepancies have been observed between different techniques for MRD assessment. In 62 patients with AML, aged 18-60 years, in first complete remission after intensive induction therapy on the randomized phase III SWOG-S0106 clinical trial (clinicaltrials gov. Identifier: NCT00085709), MRD detection by centralized, high-quality multiparametric flow cytometry was compared with a 29-gene panel utilizing duplex sequencing (DS), an ultrasensitive next-generation sequencing method that generates double-stranded consensus sequences to reduce false positive errors. MRD as defined by DS was observed in 22 (35%) patients and was strongly associated with higher rates of relapse (68% vs. 13%; hazard ratio [HR] =8.8; 95% confidence interval [CI]: 3.2-24.5; P<0.001) and decreased survival (32% vs. 82%; HR=5.6; 95% CI: 2.3-13.8; P<0.001) at 5 years. DS MRD strongly outperformed multiparametric flow cytometry MRD, which was observed in ten (16%) patients and marginally associated with higher rates of relapse (50% vs. 30%; HR=2.4; 95% CI: 0.9-6.7; P=0.087) and decreased survival (40% vs. 68%; HR=2.5; 95% CI: 1.0-6.3; P=0.059) at 5 years. Furthermore, the prognostic significance of DS MRD status at the time of remission for subsequent relapse was similar on both randomized arms of the trial. These findings suggest that next-generation sequencing-based AML MRD testing is a powerful tool that could be developed for use in patient management and for early anti-leukemic treatment assessment in clinical trials.
Collapse
Affiliation(s)
- Laura W Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Hassan Nasif
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Megan Othus
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Lan Beppu
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | | | | | - Brent L Wood
- Dept. of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | | | - Jerald P Radich
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
14
|
Falini B, Dillon R. Criteria for Diagnosis and Molecular Monitoring of NPM1-Mutated AML. Blood Cancer Discov 2024; 5:8-20. [PMID: 37917833 PMCID: PMC10772525 DOI: 10.1158/2643-3230.bcd-23-0144] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/28/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023] Open
Abstract
NPM1-mutated acute myeloid leukemia (AML) represents the largest molecular subgroup of adult AML. NPM1-mutated AML is recognizable by molecular techniques and immunohistochemistry, which, when combined, can solve difficult diagnostic problems (including identification of myeloid sarcoma and NPM1 mutations outside exon 12). According to updated 2022 European LeukemiaNet (ELN) guidelines, determining the mutational status of NPM1 (and FLT3) is a mandatory step for the genetic-based risk stratification of AML. Monitoring of measurable residual disease (MRD) by qRT-PCR, combined with ELN risk stratification, can guide therapeutic decisions at the post-remission stage. Here, we review the criteria for appropriate diagnosis and molecular monitoring of NPM1-mutated AML. SIGNIFICANCE NPM1-mutated AML represents a distinct entity in the 2022 International Consensus Classification and 5th edition of World Health Organization classifications of myeloid neoplasms. The correct diagnosis of NPM1-mutated AML and its distinction from other AML entities is extremely important because it has clinical implications for the management of AML patients, such as genetic-based risk stratification according to 2022 ELN. Monitoring of MRD by qRT-PCR, combined with ELN risk stratification, can guide therapeutic decisions at the post-remission stage, e.g., whether or not to perform allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Brunangelo Falini
- Institute of Hematology and Center for Hemato-Oncological Research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| |
Collapse
|
15
|
Thol F, Döhner H, Ganser A. How I treat refractory and relapsed acute myeloid leukemia. Blood 2024; 143:11-20. [PMID: 37944143 DOI: 10.1182/blood.2023022481] [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: 09/12/2023] [Revised: 10/11/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023] Open
Abstract
ABSTRACT Most patients with acute myeloid leukemia (AML) develop refractory/relapsed (R/R) disease even in the presence of novel and targeted therapies. Given the biological complexity of the disease and differences in frontline treatments, there are therapies approved for only subgroups of R/R AML, and enrollment in clinical trials should be first priority. Allogeneic hematopoietic cell transplantation (HCT) is the only potentially curative strategy for most patients. Therapeutic approaches, including allogeneic HCT, triggered by the presence of measurable residual disease (MRD), have recently evolved to prevent overt hematologic relapse. Salvage therapy with chemotherapy or targeted therapy is frequently administered before HCT to reduce the leukemic burden. Gilteritinib is approved by the Food and Drug Administration and European Medicines Agency for patients with relapsed FLT3 mutated AML, whereas targeted therapy for relapsed IDH1/2 mutated AML has only FDA approval. Patients who are R/R after azacitidine and venetoclax (AZA/VEN) have a dismal outcome. In this setting, even available targeted therapies show unsatisfactory results. Examples of ongoing developments include menin inhibitors, a targeted therapy for patients with mutated NPM1 or KMT2A rearrangements, antibodies targeting the macrophage immune checkpoint CD47, and triple combinations involving AZA/VEN. The latter cause significant myelosuppressive effects, which make it challenging to find the right schedule and dose.
Collapse
Affiliation(s)
- Felicitas Thol
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, 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
| |
Collapse
|
16
|
Bergeron J, Capo-Chichi JM, Tsui H, Mahe E, Berardi P, Minden MD, Brandwein JM, Schuh AC. The Clinical Utility of FLT3 Mutation Testing in Acute Leukemia: A Canadian Consensus. Curr Oncol 2023; 30:10410-10436. [PMID: 38132393 PMCID: PMC10742150 DOI: 10.3390/curroncol30120759] [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: 11/08/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) mutations are detected in approximately 20-30% of patients with acute myeloid leukemia (AML), with the presence of a FLT3 internal tandem duplication (FLT3-ITD) mutation being associated with an inferior outcome. Assessment of FLT3 mutational status is now essential to define optimal upfront treatment in both newly diagnosed and relapsed AML, to support post-induction allogeneic hematopoietic stem cell transplantation (alloSCT) decision-making, and to evaluate treatment response via measurable (minimal) residual disease (MRD) evaluation. In view of its importance in AML diagnosis and management, the Canadian Leukemia Study Group/Groupe canadien d'étude sur la leucémie (CLSG/GCEL) undertook the development of a consensus statement on the clinical utility of FLT3 mutation testing, as members reported considerable inter-center variability across Canada with respect to testing availability and timing of use, methodology, and interpretation. The CLSG/GCEL panel identified key clinical and hematopathological questions, including: (1) which patients should be tested for FLT3 mutations, and when?; (2) which is the preferred method for FLT3 mutation testing?; (3) what is the clinical relevance of FLT3-ITD size, insertion site, and number of distinct FLT3-ITDs?; (4) is there a role for FLT3 analysis in MRD assessment?; (5) what is the clinical relevance of the FLT3-ITD allelic burden?; and (6) how should results of FLT3 mutation testing be reported? The panel followed an evidence-based approach, taken together with Canadian clinical and laboratory experience and expertise, to create a consensus document to facilitate a more uniform approach to AML diagnosis and treatment across Canada.
Collapse
Affiliation(s)
- Julie Bergeron
- CEMTL Installation Maisonneuve-Rosemont, Institut Universitaire d’Hématologie-Oncologie et de Thérapie Cellulaire, Université de Montréal, Montréal, QC H1T 2M4, Canada
| | - Jose-Mario Capo-Chichi
- Division of Clinical Laboratory Genetics, Department of Laboratory Medicine and Pathobiology, Laboratory Medicine Program, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada;
| | - Hubert Tsui
- Division of Hematological Pathology, Department of Laboratory Medicine and Molecular Diagnostics, Precision Diagnostics and Therapeutics Program, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Laboratory Medicine and Pathobiology, Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Etienne Mahe
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Division of Hematology and Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Philip Berardi
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital/Eastern Ontario Regional Laboratory Association, Ottawa, ON K1H 8M2, Canada;
- Department of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mark D. Minden
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.D.M.); (A.C.S.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Joseph M. Brandwein
- Division of Hematology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada;
| | - Andre C. Schuh
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.D.M.); (A.C.S.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
17
|
Cloos J. Understanding differential technologies for detection of MRD and how to incorporate into clinical practice. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:682-690. [PMID: 38066915 PMCID: PMC10727023 DOI: 10.1182/hematology.2023000454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Patient- and leukemia-specific factors assessed at diagnosis classify patients with acute myeloid leukemia (AML) in risk categories that are prognostic for outcome. The induction phase with intensive chemotherapy in fit patients aims to reach a complete remission (CR) of less than 5% blasts in bone marrow by morphology. To deepen and sustain the response, induction is followed by consolidation treatment. This postremission treatment of patients with AML is graduated in intensity based on this favorable, intermediate, or adverse risk group classification as defined in the European Leukemia Network (ELN) 2022 recommendations. The increment of evidence that measurable residual disease (MRD) after induction can be superimposed on risk group at diagnosis is instrumental in tailoring further treatment accordingly. Several techniques are applied to detect MRD such as multiparameter flow cytometry (MFC), quantitative (digital) polymerase chain reaction (PCR), and next-generation sequencing. The clinical implementation of MRD and the technique used differ among institutes, leading to the accumulation of a wide range of data, and therefore harmonization is warranted. Currently, evidence for MRD guidance is limited to the time point after induction using MFC or quantitative PCR for NPM1 and core binding factor abnormalities in intermediate-risk patients. The role of MRD in targeted or nonintensive therapies needs to be clarified, although some data show improved survival in patients achieving CR-MRD negativity. Potential application of MRD for selection of conditioning before stem cell transplantation, monitoring after consolidation, and use as an intermediate end point in clinical trials need further evaluation.
Collapse
Affiliation(s)
- Jacqueline Cloos
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, location VUMC, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Loo S, Roberts AW, Anstee NS, Kennedy GA, He S, Schwarer AP, Enjeti AK, D’Rozario J, Marlton P, Bilmon IA, Taper J, Cull G, Tiley C, Verner E, Hahn U, Hiwase DK, Iland HJ, Murphy N, Ramanathan S, Reynolds J, Ong DM, Tiong IS, Wall M, Murray M, Rawling T, Leadbetter J, Rowley L, Latimer M, Yuen S, Ting SB, Fong CY, Morris K, Bajel A, Seymour JF, Levis MJ, Wei AH. Sorafenib plus intensive chemotherapy in newly diagnosed FLT3-ITD AML: a randomized, placebo-controlled study by the ALLG. Blood 2023; 142:1960-1971. [PMID: 37647654 PMCID: PMC10733823 DOI: 10.1182/blood.2023020301] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 09/01/2023] Open
Abstract
Sorafenib maintenance improves outcomes after hematopoietic cell transplant (HCT) for patients with FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) acute myeloid leukemia (AML). Although promising outcomes have been reported for sorafenib plus intensive chemotherapy, randomized data are limited. This placebo-controlled, phase 2 study (ACTRN12611001112954) randomized 102 patients (aged 18-65 years) 2:1 to sorafenib vs placebo (days 4-10) combined with intensive induction: idarubicin 12 mg/m2 on days 1 to 3 plus either cytarabine 1.5 g/m2 twice daily on days 1, 3, 5, and 7 (18-55 years) or 100 mg/m2 on days 1 to 7 (56-65 years), followed by consolidation and maintenance therapy for 12 months (post-HCT excluded) in newly diagnosed patients with FLT3-ITD AML. Four patients were excluded in a modified intention-to-treat final analysis (3 not commencing therapy and 1 was FLT3-ITD negative). Rates of complete remission (CR)/CR with incomplete hematologic recovery were high in both arms (sorafenib, 78%/9%; placebo, 70%/24%). With 49.1-months median follow-up, the primary end point of event-free survival (EFS) was not improved by sorafenib (2-year EFS 47.9% vs 45.4%; hazard ratio [HR], 0.87; 95% confidence interval [CI], 0.51-1.51; P = .61). Two-year overall survival (OS) was 67% in the sorafenib arm and 58% in the placebo arm (HR, 0.76; 95% CI, 0.42-1.39). For patients who received HCT in first remission, the 2-year OS rates were 84% and 67% in the sorafenib and placebo arms, respectively (HR, 0.45; 95% CI, 0.18-1.12; P = .08). In exploratory analyses, FLT3-ITD measurable residual disease (MRD) negative status (<0.001%) after induction was associated with improved 2-year OS (83% vs 60%; HR, 0.4; 95% CI, 0.17-0.93; P = .028). In conclusion, routine use of pretransplant sorafenib plus chemotherapy in unselected patients with FLT3-ITD AML is not supported by this study.
Collapse
Affiliation(s)
- Sun Loo
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
| | - Andrew W. Roberts
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
| | - Natasha S. Anstee
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
| | - Glen A. Kennedy
- Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
| | - Simon He
- Department of Clinical Haematology, Austin Health, Heidelberg, VIC, Australia
| | | | - Anoop K. Enjeti
- Calvary Mater Newcastle Hospital, Waratah, NSW, Australia
- University of Newcastle, Callaghan, NSW, Australia
| | | | - Paula Marlton
- Princess Alexandra Hospital and University of Queensland, Woolloongabba, QLD, Australia
| | - Ian A. Bilmon
- Department of Haematology, Westmead Hospital, Westmead, NSW, Australia
| | - John Taper
- Nepean Hospital Cancer Care Centre, Kingswood, NSW, Australia
| | - Gavin Cull
- Sir Charles Gairdner Hospital, University of Western Australia, Crawley, WA, Australia
| | | | - Emma Verner
- Concord Repatriation General Hospital, Concord, NSW, Australia
| | - Uwe Hahn
- Department of Haematology, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Devendra K. Hiwase
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Harry J. Iland
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- University of Sydney, Camperdown, NSW, Australia
| | - Nick Murphy
- Royal Hobart Hospital, Hobart, TS, Australia
| | | | - John Reynolds
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Doen Ming Ong
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Ing Soo Tiong
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Meaghan Wall
- Murdoch Children’s Research Institute, Melbourne, VIC, Australia
| | - Michael Murray
- Sydney Pharmacy School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | | | | | - Leesa Rowley
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC, Australia
| | | | - Sam Yuen
- Calvary Mater Newcastle Hospital, Waratah, NSW, Australia
| | - Stephen B. Ting
- Department of Haematology, Box Hill Hospital, Box Hill, VIC, Australia
| | - Chun Yew Fong
- Department of Clinical Haematology, Austin Health, Heidelberg, VIC, Australia
| | - Kirk Morris
- Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
| | - Ashish Bajel
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - John F. Seymour
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Mark J. Levis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Andrew H. Wei
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Australasian Leukaemia and Lymphoma Group
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
- Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
- Department of Clinical Haematology, Austin Health, Heidelberg, VIC, Australia
- Department of Haematology, Box Hill Hospital, Box Hill, VIC, Australia
- Calvary Mater Newcastle Hospital, Waratah, NSW, Australia
- University of Newcastle, Callaghan, NSW, Australia
- Canberra Hospital, Garran, ACT, Australia
- Princess Alexandra Hospital and University of Queensland, Woolloongabba, QLD, Australia
- Department of Haematology, Westmead Hospital, Westmead, NSW, Australia
- Nepean Hospital Cancer Care Centre, Kingswood, NSW, Australia
- Sir Charles Gairdner Hospital, University of Western Australia, Crawley, WA, Australia
- Gosford Hospital, Gosford, NSW, Australia
- Concord Repatriation General Hospital, Concord, NSW, Australia
- Department of Haematology, The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- University of Sydney, Camperdown, NSW, Australia
- Royal Hobart Hospital, Hobart, TS, Australia
- St George Hospital, Kogarah, NSW, Australia
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, VIC, Australia
- Murdoch Children’s Research Institute, Melbourne, VIC, Australia
- Sydney Pharmacy School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- University of Technology Sydney, Sydney, NSW, Australia
- WriteSource Medical Pty Ltd, Lane Cove, NSW, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC, Australia
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
19
|
Stone RM. FLT(3)-ing about: the search for the best inhibitor. Blood 2023; 142:1937-1938. [PMID: 38060271 DOI: 10.1182/blood.2023022174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
|
20
|
Ravandi F, Cloos J, Buccisano F, Dillon R, Döhner K, Freeman SD, Hourigan CS, Ossenkoppele GJ, Roboz GJ, Subklewe M, Thiede C, Arnhardt I, Valk PJM, Venditti A, Wei AH, Walter RB, Heuser M. Measurable residual disease monitoring in patients with acute myeloid leukemia treated with lower-intensity therapy: Roadmap from an ELN-DAVID expert panel. Am J Hematol 2023; 98:1847-1855. [PMID: 37671649 PMCID: PMC10841357 DOI: 10.1002/ajh.27087] [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: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/07/2023]
Abstract
With the availability of effective targeted agents, significant changes have occurred in the management of patients with acute myeloid leukemia (AML) over the past several years, particularly for those considered unfit for intensive chemotherapy. While testing for measurable residual disease (MRD) is now routinely performed in patients treated with intensive chemotherapy to refine prognosis and, possibly, inform treatment decision-making, its value in the context of lower-intensity regimens is unclear. As such regimens have gained in popularity and can be associated with higher response rates, the need to better define the role of MRD assessment and the appropriate time points and assays used for this purpose has increased. This report outlines a roadmap for MRD testing in patients with AML treated with lower-intensity regimens. Experts from the European LeukemiaNet (ELN)-DAVID AML MRD working group reviewed all available data to propose a framework for MRD testing in future trials and clinical practice. A Delphi poll served to optimize consensus. Establishment of uniform standards for MRD assessments in lower-intensity regimens used in treating patients with AML is clinically relevant and important for optimizing testing and, ultimately, improving treatment outcomes of these patients.
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, The University of Texas - MD Anderson Cancer Center, Houston
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, Hematology, University Tor Vergata, Rome, Italy
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King’s College, London, United Kingdom
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Sylvie D Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancy, Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda
| | - Gerrit J Ossenkoppele
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gail J Roboz
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine, New York
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University Munich, Germany
| | - Christian Thiede
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Isabell Arnhardt
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Peter J M Valk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Adriano Venditti
- Department of Biomedicine and Prevention, Hematology, University Tor Vergata, Rome, Italy
| | - Andrew H Wei
- Department of Haematology, Peter MacCallum Cancer Centre, Royal Melbourne Hospital, University of Melbourne and Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Roland B Walter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
21
|
Hokland P, Fernández II, Freeman SD, Gjertsen BT, Jin J, Murthy V, Yanada M, Ganser A. AML in the elderly-A global view. Br J Haematol 2023; 203:760-773. [PMID: 37822071 DOI: 10.1111/bjh.19135] [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: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023]
Affiliation(s)
- Peter Hokland
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Isolda I Fernández
- Fundación para Combatir la Leucemia, Department of Hematology, Buenos Aires, Argentina
| | - Sylvie D Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Bjørn T Gjertsen
- Department of Clinical Science, Haukeland Centre for Cancer Biomarkers (CCBIO), University of Bergen, Helse Bergen HF, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Helse Bergen HF, Bergen, Norway
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Vidhya Murthy
- Centre for Clinical Haematology, University Hospitals Birmingham, Birmingham, UK
| | - Masamitsu Yanada
- Department of Hematology and Oncology, Nagoya City University East Medical Center, Nagoya, Japan
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
22
|
Blackmon AL, Hourigan CS. Test Then Erase? Current Status and Future Opportunities for Measurable Residual Disease Testing in Acute Myeloid Leukemia. Acta Haematol 2023; 147:133-146. [PMID: 38035547 PMCID: PMC10963159 DOI: 10.1159/000535463] [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: 08/17/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Measurable residual disease (MRD) test positivity during and after treatment in patients with acute myeloid leukemia (AML) has been associated with higher rates of relapse and worse overall survival. Current approaches for MRD testing are not standardized leading to inconsistent results and poor prognostication of disease. Pertinent studies evaluating AML MRD testing at specific times points, with various therapeutics and testing methods are presented. SUMMARY AML is a set of diseases with different molecular and cytogenetic characteristics and is often polyclonal with evolution over time. This genetic diversity poses a great challenge for a single AML MRD testing approach. The current ELN 2021 MRD guidelines recommend MRD testing by quantitative polymerase chain reaction in those with a validated molecular target or multiparameter flow cytometry (MFC) in all other cases. The benefit of MFC is the ability to use this method across disease subsets, at the relative expense of suboptimal sensitivity and specificity. AML MRD detection may be improved with molecular methods. Genetic characterization at AML diagnosis and relapse is now standard of care for appropriate therapeutic assignment, and future initiatives will provide the evidence to support testing in remission to direct clinical interventions. KEY MESSAGES The treatment options for patients with AML have expanded for specific molecular subsets such as FLT3 and IDH1/2 mutated AML, with development of novel agents for NPM1 mutated or KMT2A rearranged AML ongoing, but also due to effective venetoclax-combinations. Evidence regarding highly sensitive molecular MRD detection methods for specific molecular subgroups, in the context of these new treatment approaches, will likely shape the future of AML care.
Collapse
Affiliation(s)
- Amanda L. Blackmon
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Christopher S. Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
23
|
Harrop S, Nguyen PC, Byrne D, Wilson C, Ryland GL, Nguyen T, Anderson MA, Khaw SL, Martin M, Tiong IS, Sanij E, Blombery P. Persistence of UBTF tandem duplications in remission in acute myeloid leukaemia. EJHAEM 2023; 4:1105-1109. [PMID: 38024622 PMCID: PMC10660390 DOI: 10.1002/jha2.808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023]
Abstract
UBTF tandem duplications are recurrent in adult and paediatric acute myeloid leukaemia and have been reported to be associated with a poor prognosis. Co-mutations in WT1 and FLT3 are common while morphological dysplasia is frequent. The role of UBTF-TDs in leukemogenesis is yet to be elucidated; however they have been proposed as early initiating events, making them attractive for assessment of MRD and a potential therapeutic target. We present two cases where the UBTF-TD was observed in remission and discuss the implications of these findings in the clinicobiological understanding of this emerging entity.
Collapse
Affiliation(s)
- Sean Harrop
- Peter MacCallum Cancer Centre Melbourne Victoria Australia
| | | | - David Byrne
- Peter MacCallum Cancer Centre Melbourne Victoria Australia
| | | | - Georgina L Ryland
- Peter MacCallum Cancer Centre Melbourne Victoria Australia
- Sir Peter MacCallum Department of Oncology University of Melbourne Parkville Victoria Australia
| | - Tamia Nguyen
- Peter MacCallum Cancer Centre Melbourne Victoria Australia
| | | | - Seong Lin Khaw
- Royal Children's Hospital Melbourne Victoria Australia
- Murdoch Children's Research Institute Melbourne Victoria Australia
| | | | - Ing Soo Tiong
- Peter MacCallum Cancer Centre Melbourne Victoria Australia
| | - Elaine Sanij
- Peter MacCallum Cancer Centre Melbourne Victoria Australia
- St Vincent's Institute of Medical Research Fitzroy Victoria Australia
- Department of Medicine St Vincent's Hospital University of Melbourne Melbourne Victoria Australia
- Sir Peter MacCallum Department of Oncology University of Melbourne Parkville Victoria Australia
- Department of Biochemistry and Molecular Biology Monash University Clayton Victoria Australia
| | - Piers Blombery
- Peter MacCallum Cancer Centre Melbourne Victoria Australia
- Sir Peter MacCallum Department of Oncology University of Melbourne Parkville Victoria Australia
| |
Collapse
|
24
|
Hernández-Sánchez A, Bullinger L. Recent advances in precision medicine for acute myeloid leukemia. Curr Opin Oncol 2023; 35:581-588. [PMID: 37621173 DOI: 10.1097/cco.0000000000000965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
PURPOSE OF REVIEW Acute myeloid leukemia (AML) is a heterogeneous disease, in which treatment response and patient survival are highly conditioned by the leukemia biology. The aim of this review is to summarize recent advances in AML classification, risk stratification models, measurable residual disease (MRD) and the increasing number of treatment options that are paving the way towards precision medicine in AML. RECENT FINDINGS AML classification and risk stratification were recently updated by incorporating novel molecular markers that are important for diagnosis and outcome prediction. In addition, the impact of co-mutational patterns is under investigation and novel approaches using machine learning algorithms are starting to be used for individualized risk estimation. Molecular markers are also becoming useful in predicting response to non-intensive treatments. MRD informs of treatment response with high sensitivity, allowing dynamic patient risk assessment and early intervention. Finally, important advances were made in AML therapy, with an increasing number of targeted therapies becoming available and many novel treatment approaches being under development with promising early results. SUMMARY A better understanding of AML biology is leading to improved risk stratification and important advances in treatments, which are allowing the development of precision medicine in AML at an unprecedented pace.
Collapse
Affiliation(s)
- Alberto Hernández-Sánchez
- University Hospital of Salamanca
- Institute of Biomedical Research of Salamanca (IBSAL) , Salamanca, Spain
| | | |
Collapse
|
25
|
Othman J, Potter N, Mokretar K, Taussig D, Khan A, Krishnamurthy P, Latif AL, Cahalin P, Aries J, Amer M, Belsham E, Conneally E, Craddock C, Culligan D, Dennis M, Duncan C, Freeman SD, Furness C, Gilkes A, Gkreka P, Hodgson K, Ingram W, Jain M, King A, Knapper S, Kottaridis P, McMullin MF, Mohite U, Ngu L, O'Nions J, Patrick K, Rider T, Roberts W, Severinsen MT, Storrar N, Taylor T, Russell NH, Dillon R. FLT3 inhibitors as MRD-guided salvage treatment for molecular failure in FLT3 mutated AML. Leukemia 2023; 37:2066-2072. [PMID: 37558736 PMCID: PMC10539160 DOI: 10.1038/s41375-023-01994-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/17/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
Patients with FLT3-mutated AML have a high relapse rate and suboptimal outcomes. Many have co-mutations suitable for measurable residual disease (MRD) monitoring by RT-qPCR and those destined to relapse can be identified by high or rising levels of MRD, called molecular failure. This provides a window for pre-emptive intervention, but there is little evidence to guide treatment. The use of FLT3 inhibitors (FLT3i) appears attractive but their use has not yet been evaluated. We identified 56 patients treated with FLT3i at molecular failure. The FLT3 mutation was an ITD in 52, TKD in 7 and both in 3. Over half of patients had previously received midostaurin. Molecular failure occurred at a median 9.2 months from diagnosis and was treated with gilteritinib (n = 38), quizartinib (n = 7) or sorafenib (n = 11). 60% achieved a molecular response, with 45% reaching MRD negativity. Haematological toxicity was low, and 22 patients were bridged directly to allogeneic transplant with another 6 to donor lymphocyte infusion. 2-year overall survival was 80% (95%CI 69-93) and molecular event-free survival 56% (95%CI 44-72). High-sensitivity next-generation sequencing for FLT3-ITD at molecular failure identified patients more likely to benefit. FLT3i monotherapy for molecular failure is a promising strategy which merits evaluation in prospective studies.
Collapse
Affiliation(s)
- Jad Othman
- Department of Medical and Molecular Genetics, King's College London, London, England, UK
- Guy's and St Thomas' NHS Foundation Trust, London, England, UK
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Nicola Potter
- Department of Medical and Molecular Genetics, King's College London, London, England, UK
| | | | - David Taussig
- The Royal Marsden NHS Foundation Trust, London, England, UK
| | - Anjum Khan
- Leeds Teaching Hospitals NHS Trust, Leeds, England, UK
| | | | | | - Paul Cahalin
- Blackpool Teaching Hospitals NHS Foundation Trust, Blackpool, England, UK
| | - James Aries
- Barts Cancer Institute, Queen Mary University of London, London, England, UK
| | - Mariam Amer
- University Hospital Southampton, Southampton, England, UK
| | | | | | | | | | - Mike Dennis
- The Christie NHS Foundation Trust, Manchester, England, UK
| | | | - Sylvie D Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, Scotland, UK
| | | | - Amanda Gilkes
- Department of Haematology, Cardiff University, Cardiff, Wales, UK
| | | | | | | | - Manish Jain
- Leeds Teaching Hospitals NHS Trust, Leeds, England, UK
| | - Andrew King
- Addenbrooke's Hospital, Cambridge, England, UK
| | - Steven Knapper
- School of Medicine, Cardiff University, Cardiff, Wales, UK
| | | | | | | | - Loretta Ngu
- Royal Devon & Exeter NHS Foundation Trust, Exeter, England, UK
| | - Jenny O'Nions
- University College London Hospital NHS Foundation Trust, London, England, UK
| | | | - Tom Rider
- The Royal Sussex County Hospital, Brighton and Hove, England, UK
| | - Wing Roberts
- Great North Children's Hospital, Newcastle upon Tyne, England, UK
| | - Marianne Tang Severinsen
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | | | - Tom Taylor
- Nottingham University Hospital, Nottingham, England, UK
| | - Nigel H Russell
- Guy's and St Thomas' NHS Foundation Trust, London, England, UK
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College London, London, England, UK.
- Guy's and St Thomas' NHS Foundation Trust, London, England, UK.
| |
Collapse
|
26
|
Swaminathan M, Ravandi F. Can measurable residual disease assessment be reliably used to defer allogeneic stem cell transplant in patients with intermediate-risk acute myeloid leukemia? Haematologica 2023; 108:2561-2563. [PMID: 37345488 PMCID: PMC10543186 DOI: 10.3324/haematol.2023.283120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023] Open
Affiliation(s)
- Mahesh Swaminathan
- The University of Texas, MD Anderson Cancer Center, Department of Leukemia, Texas
| | - Farhad Ravandi
- The University of Texas, MD Anderson Cancer Center, Department of Leukemia, Texas.
| |
Collapse
|
27
|
Madaci L, Farnault L, Abbou N, Gabert J, Venton G, Costello R. Impact of Next-Generation Sequencing in Diagnosis, Prognosis and Therapeutic Management of Acute Myeloid Leukemia/Myelodysplastic Neoplasms. Cancers (Basel) 2023; 15:3280. [PMID: 37444390 DOI: 10.3390/cancers15133280] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
For decades, the diagnosis, prognosis and thus, the treatment of acute myeloblastic leukemias and myelodysplastic neoplasms has been mainly based on morphological aspects, as evidenced by the French-American-British classification. The morphological aspects correspond quite well, in a certain number of particular cases, to particular evolutionary properties, such as acute myelomonoblastic leukemias with eosinophils or acute promyelocytic leukemias. Advances in biology, particularly "classical" cytogenetics (karyotype) and molecular cytogenetics (in situ hybridization), have made it possible to associate certain morphological features with particular molecular abnormalities, such as the pericentric inversion of chromosome 16 and translocation t(15;17) in the two preceding examples. Polymerase chain reaction techniques have made it possible to go further in these analyses by associating these karyotype abnormalities with their molecular causes, CBFbeta fusion with MYH11 and PML-RAR fusion in the previous cases. In these two examples, the molecular abnormality allows us to better define the pathophysiology of leukemia, to adapt certain treatments (all-transretinoic acid, for example), and to follow up the residual disease of strong prognostic value beyond the simple threshold of less than 5% of marrow blasts, signaling the complete remission. However, the new sequencing techniques of the next generation open up broader perspectives by being able to analyze several dozens of molecular abnormalities, improving all levels of management, from diagnosis to prognosis and treatment, even if it means that morphological aspects are increasingly relegated to the background.
Collapse
Affiliation(s)
- Lamia Madaci
- TAGC, INSERM, UMR1090, Aix-Marseille University, 13005 Marseille, France
| | - Laure Farnault
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
| | - Norman Abbou
- Molecular Biology Laboratory, Timone University Hospital, 13005 Marseille, France
| | - Jean Gabert
- Molecular Biology Laboratory, Timone University Hospital, 13005 Marseille, France
| | - Geoffroy Venton
- TAGC, INSERM, UMR1090, Aix-Marseille University, 13005 Marseille, France
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
| | - Régis Costello
- TAGC, INSERM, UMR1090, Aix-Marseille University, 13005 Marseille, France
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
| |
Collapse
|
28
|
Wong ZC, Dillon LW, Hourigan CS. Measurable residual disease in patients undergoing allogeneic transplant for acute myeloid leukemia. Best Pract Res Clin Haematol 2023; 36:101468. [PMID: 37353292 PMCID: PMC10291441 DOI: 10.1016/j.beha.2023.101468] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/11/2023] [Indexed: 06/25/2023]
Abstract
The most common indication for allogeneic hematopoietic cell transplant (alloHCT) is maintenance of remission after initial treatment for patients with acute myeloid leukemia (AML). Loss of remission, relapse, remains however the most frequent cause of alloHCT failure. There is strong evidence that detectable persistent disease burden ("measurable residual disease", MRD) in patients with AML in remission prior to alloHCT is associated with increased risk of post-transplant relapse. MRD status as a summative assessment of response to pre-transplant therapy may allow superior patient-personalized risk stratification compared with models solely incorporating pre-treatment variables. An optimal methodology for AML MRD detection has not yet been established, but molecular methods such as DNA-sequencing may have additional prognostic utility compared to current approaches. There is growing evidence that intervention on AML MRD positivity may improve post-transplant outcomes. New initiatives will generate actionable data on the clinical utility of AML MRD testing for patients undergoing alloHCT.
Collapse
Affiliation(s)
- Zoë C Wong
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD, USA
| | - Laura W Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD, USA
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
29
|
Dekker SE, Rea D, Cayuela JM, Arnhardt I, Leonard J, Heuser M. Using Measurable Residual Disease to Optimize Management of AML, ALL, and Chronic Myeloid Leukemia. Am Soc Clin Oncol Educ Book 2023; 43:e390010. [PMID: 37311155 DOI: 10.1200/edbk_390010] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In this review, we discuss the use of measurable residual disease (MRD) in AML, ALL, and chronic myeloid leukemia (CML). Our aims were to review the different methodologies for MRD assessment; describe the clinical relevance and medical decision making on the basis of MRD; compare and contrast the usage of MRD across AML, ALL, and CML; and discuss what patients need to know about MRD as it relates to their disease status and treatment. Finally, we discuss ongoing challenges and future directions with the goal of optimizing MRD usage in leukemia management.
Collapse
Affiliation(s)
- Simone E Dekker
- Department of Medicine, Oregon Health & Science University, Portland, OR
| | - Delphine Rea
- France Intergroupe des Leucémies Myéloïdes chroniques FiLMC, Hôpital Saint-Louis APHP, Paris, France
- Service d'Hématologie Adulte, Hôpital Saint-Louis APHP, Paris, France
| | - Jean-Michel Cayuela
- France Intergroupe des Leucémies Myéloïdes chroniques FiLMC, Hôpital Saint-Louis APHP, Paris, France
- Laboratoire de Biologie Moléculaire, Hôpital Saint-Louis APHP, Paris, France
| | - Isabell Arnhardt
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Jessica Leonard
- Division of Hematology-Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- Comprehensive Cancer Center Lower Saxony, Hannover, Germany
| |
Collapse
|
30
|
Ogbue O, Unlu S, Ibodeng GO, Singh A, Durmaz A, Visconte V, Molina JC. Single-Cell Next-Generation Sequencing to Monitor Hematopoietic Stem-Cell Transplantation: Current Applications and Future Perspectives. Cancers (Basel) 2023; 15:cancers15092477. [PMID: 37173944 PMCID: PMC10177286 DOI: 10.3390/cancers15092477] [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: 04/02/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) are genetically complex and diverse diseases. Such complexity makes challenging the monitoring of response to treatment. Measurable residual disease (MRD) assessment is a powerful tool for monitoring response and guiding therapeutic interventions. This is accomplished through targeted next-generation sequencing (NGS), as well as polymerase chain reaction and multiparameter flow cytometry, to detect genomic aberrations at a previously challenging leukemic cell concentration. A major shortcoming of NGS techniques is the inability to discriminate nonleukemic clonal hematopoiesis. In addition, risk assessment and prognostication become more complicated after hematopoietic stem-cell transplantation (HSCT) due to genotypic drift. To address this, newer sequencing techniques have been developed, leading to more prospective and randomized clinical trials aiming to demonstrate the prognostic utility of single-cell next-generation sequencing in predicting patient outcomes following HSCT. This review discusses the use of single-cell DNA genomics in MRD assessment for AML/MDS, with an emphasis on the HSCT time period, including the challenges with current technologies. We also touch on the potential benefits of single-cell RNA sequencing and analysis of accessible chromatin, which generate high-dimensional data at the cellular resolution for investigational purposes, but not currently used in the clinical setting.
Collapse
Affiliation(s)
- Olisaemeka Ogbue
- Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH 44111, USA
| | - Serhan Unlu
- Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH 44111, USA
| | - Gogo-Ogute Ibodeng
- Internal Medicine, Infirmary Health's Thomas Hospital, Fairhope, AL 36607, USA
| | - Abhay Singh
- Department of Hematology Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Arda Durmaz
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Taussig Cancer Center, Cleveland, OH 44106, USA
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Taussig Cancer Center, Cleveland, OH 44106, USA
| | - John C Molina
- Department of Hematology Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH 44106, USA
| |
Collapse
|
31
|
Yokoyama S, Onozawa M, Yoshida S, Miyashita N, Kimura H, Takahashi S, Matsukawa T, Goto H, Fujisawa S, Miki K, Hidaka D, Hashiguchi J, Wakasa K, Ibata M, Takeda Y, Shigematsu A, Fujimoto K, Tsutsumi Y, Mori A, Ishihara T, Kakinoki Y, Kondo T, Hashimoto D, Teshima T. Subclinical minute FLT3-ITD clone can be detected in clinically FLT3-ITD-negative acute myeloid leukaemia at diagnosis. Br J Haematol 2023. [PMID: 37067758 DOI: 10.1111/bjh.18800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/18/2023]
Abstract
Recent advances in next-generation sequencing (NGS) have enabled the detection of subclinical minute FLT3-ITD. We selected 74 newly diagnosed, cytogenetically normal acute myeloid leukaemia (AML) samples in which FLT3-ITD was not detected by gel electrophoresis. We sequenced them using NGS and found minute FLT3-ITDs in 19 cases. We compared cases with clinically relevant FLT3-ITD (n = 37), cases with minute FLT3-ITD (n = 19) and cases without detectable FLT3-ITD (n = 55). Molecular characteristics (location and length) of minute FLT3-ITD were similar to those of clinically relevant FLT3-ITD. Survival of cases with minute FLT3-ITD was similar to that of cases without detectable FLT3-ITD, whereas the relapse rate within 1 year after onset was significantly higher in cases with minute FLT3-ITD. We followed 18 relapsed samples of cases with clinically FLT3-ITD-negative at diagnosis. Two of 3 cases with minute FLT3-ITD relapsed with progression to clinically relevant FLT3-ITD. Two of 15 cases in which FLT3-ITD was not detected by NGS relapsed with the emergence of minute FLT3-ITD, and one of them showed progression to clinically relevant FLT3-ITD at the second relapse. We revealed the clonal dynamics of subclinical minute FLT3-ITD in clinically FLT3-ITD-negative AML. Minute FLT3-ITD at the initial AML can expand to become a dominant clone at relapse.
Collapse
Affiliation(s)
- Shota Yokoyama
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Masahiro Onozawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Shota Yoshida
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Naoki Miyashita
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Hiroyuki Kimura
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Shogo Takahashi
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Toshihiro Matsukawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Hideki Goto
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Shinichi Fujisawa
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Kosuke Miki
- Department of Hematology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Daisuke Hidaka
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | | | - Kentaro Wakasa
- Division of Hematology, Obihiro-Kosei General Hospital, Obihiro, Japan
| | - Makoto Ibata
- Department of Hematology, Sapporo-Kosei General Hospital, Sapporo, Japan
| | - Yukari Takeda
- Department of Hematology, Tonan Hospital, Sapporo, Japan
| | - Akio Shigematsu
- Department of Hematology, Kushiro Rosai Hospital, Kushiro, Japan
| | - Katsuya Fujimoto
- Department of Hematology, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | - Yutaka Tsutsumi
- Department of Hematology, Hakodate Municipal Hospital, Hakodate, Japan
| | - Akio Mori
- Blood Disorders Center, Aiiku Hospital, Sapporo, Japan
| | | | | | - Takeshi Kondo
- Blood Disorders Center, Aiiku Hospital, Sapporo, Japan
| | - Daigo Hashimoto
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
32
|
Fedorov K, Maiti A, Konopleva M. Targeting FLT3 Mutation in Acute Myeloid Leukemia: Current Strategies and Future Directions. Cancers (Basel) 2023; 15:cancers15082312. [PMID: 37190240 DOI: 10.3390/cancers15082312] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
FLT3 mutations are present in 30% of newly diagnosed patients with acute myeloid leukemia. Two broad categories of FLT3 mutations are ITD and TKD, with the former having substantial clinical significance. Patients with FLT3-ITD mutation present with a higher disease burden and have inferior overall survival, due to high relapse rates after achieving remission. The development of targeted therapies with FLT3 inhibitors over the past decade has substantially improved clinical outcomes. Currently, two FLT3 inhibitors are approved for use in patients with acute myeloid leukemia: midostaurin in the frontline setting, in combination with intensive chemotherapy; and gilteritinib as monotherapy in the relapsed refractory setting. The addition of FLT3 inhibitors to hypomethylating agents and venetoclax offers superior responses in several completed and ongoing studies, with encouraging preliminary data. However, responses to FLT3 inhibitors are of limited duration due to the emergence of resistance. A protective environment within the bone marrow makes eradication of FLT3mut leukemic cells difficult, while prior exposure to FLT3 inhibitors leads to the development of alternative FLT3 mutations as well as activating mutations in downstream signaling, promoting resistance to currently available therapies. Multiple novel therapeutic strategies are under investigation, including BCL-2, menin, and MERTK inhibitors, as well as FLT3-directed BiTEs and CAR-T therapy.
Collapse
Affiliation(s)
- Kateryna Fedorov
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marina Konopleva
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| |
Collapse
|
33
|
Dillon LW, Higgins J, Nasif H, Othus M, Beppu L, Smith TH, Schmidt E, Valentine CC, Salk JJ, Wood BL, Erba HP, Radich JP, Hourigan CS. Quantification of measurable residual disease using duplex sequencing in adults with acute myeloid leukemia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.26.23287367. [PMID: 37034683 PMCID: PMC10081409 DOI: 10.1101/2023.03.26.23287367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The presence of measurable residual disease (MRD) is strongly associated with treatment outcomes in acute myeloid leukemia (AML). Despite the correlation with clinical outcomes, MRD assessment has yet to be standardized or routinely incorporated into clinical trials. Discrepancies have been observed between different techniques for MRD assessment and there remains a need to compare centralized, high-quality multiparametric flow cytometry (MFC) and ultrasensitive next-generation sequencing (NGS) in AML patients with diverse mutational profiles. In 62 patients with AML, aged 18-60, in first complete remission after intensive induction therapy on the randomized phase 3 SWOG-S0106 clinical trial, MRD detection by MFC was compared with a 29 gene panel utilizing duplex sequencing (DS), an NGS method that generates double-stranded consensus sequences to reduce false positive errors. Using DS, detection of a persistent mutation utilizing defined criteria was seen in 22 (35%) patients and was strongly associated with higher rates of relapse (68% vs 13% at year 5; HR, 8.8; 95% CI, 3.2-24.5; P<0.001) and decreased survival (32% vs 82% at year 5; HR, 5.6; 95% CI, 2.3-13.8; P<0.001). MRD as defined by DS strongly outperformed MFC, which was observed in 10 (16%) patients and marginally associated with higher rates of relapse (50% vs 30% at year 5; HR, 2.4; 95% CI, 0.9-6.7; P=0.087) and decreased survival (40% vs 68% at year 5; HR, 2.5; 95% CI, 1.0-6.3; P=0.059). Furthermore, the prognostic significance of DS MRD status at the time of remission was similar on both randomized arms of the trial, predicting S0106 clinical trial outcomes. These findings suggest that DS is a powerful tool that could be used in patient management and for early treatment assessment in clinical trials.
Collapse
Affiliation(s)
- Laura W. Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Hassan Nasif
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Megan Othus
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Lan Beppu
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | | | | | - Brent L Wood
- Dept. of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA
| | | | - Jerald P. Radich
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Christopher S. Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD
| |
Collapse
|
34
|
Bhansali RS, Pratz KW, Lai C. Recent advances in targeted therapies in acute myeloid leukemia. J Hematol Oncol 2023; 16:29. [PMID: 36966300 PMCID: PMC10039574 DOI: 10.1186/s13045-023-01424-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/14/2023] [Indexed: 03/27/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. While survival for younger patients over the last several decades has improved nearly sixfold with the optimization of intensive induction chemotherapy and allogeneic stem cell transplantation (alloHSCT), this effect has been largely mitigated in older and less fit patients as well as those with adverse-risk disease characteristics. However, the last 10 years has been marked by major advances in the molecular profiling of AML characterized by a deeper understanding of disease pathobiology and therapeutic vulnerabilities. In this regard, the classification of AML subtypes has recently evolved from a morphologic to a molecular and genetic basis, reflected by recent updates from the World Health Organization and the new International Consensus Classification system. After years of stagnation in new drug approvals for AML, there has been a rapid expansion of the armamentarium against this disease since 2017. Low-intensity induction therapy with hypomethylating agents and venetoclax has substantially improved outcomes, including in those previously considered to have a poor prognosis. Furthermore, targeted oral therapies against driver mutations in AML have been added to the repertoire. But with an accelerated increase in treatment options, several questions arise such as how to best sequence therapy, how to combine therapies, and if there is a role for maintenance therapy in those who achieve remission and cannot undergo alloHSCT. Moreover, certain subtypes of AML, such as those with TP53 mutations, still have dismal outcomes despite these recent advances, underscoring an ongoing unmet need and opportunity for translational advances. In this review, we will discuss recent updates in the classification and risk stratification of AML, explore the literature regarding low-intensity and novel oral combination therapies, and briefly highlight investigative agents currently in early clinical development for high-risk disease subtypes.
Collapse
Affiliation(s)
- Rahul S Bhansali
- Division of Hematology/Oncology, Department of Medicine, Hospital of the University of Pennsylvania, South Pavilion, 12th Floor, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Keith W Pratz
- Division of Hematology/Oncology, Department of Medicine, Hospital of the University of Pennsylvania, South Pavilion, 12th Floor, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Catherine Lai
- Division of Hematology/Oncology, Department of Medicine, Hospital of the University of Pennsylvania, South Pavilion, 12th Floor, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
35
|
Dillon LW, Gui G, Page KM, Ravindra N, Wong ZC, Andrew G, Mukherjee D, Zeger SL, El Chaer F, Spellman S, Howard A, Chen K, Auletta J, Devine SM, Jimenez Jimenez AM, De Lima MJG, Litzow MR, Kebriaei P, Saber W, Weisdorf DJ, Hourigan CS. DNA Sequencing to Detect Residual Disease in Adults With Acute Myeloid Leukemia Prior to Hematopoietic Cell Transplant. JAMA 2023; 329:745-755. [PMID: 36881031 PMCID: PMC9993183 DOI: 10.1001/jama.2023.1363] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/28/2023] [Indexed: 03/08/2023]
Abstract
Importance Preventing relapse for adults with acute myeloid leukemia (AML) in first remission is the most common indication for allogeneic hematopoietic cell transplant. The presence of AML measurable residual disease (MRD) has been associated with higher relapse rates, but testing is not standardized. Objective To determine whether DNA sequencing to identify residual variants in the blood of adults with AML in first remission before allogeneic hematopoietic cell transplant identifies patients at increased risk of relapse and poorer overall survival compared with those without these DNA variants. Design, Setting, and Participants In this retrospective observational study, DNA sequencing was performed on pretransplant blood from patients aged 18 years or older who had undergone their first allogeneic hematopoietic cell transplant during first remission for AML associated with variants in FLT3, NPM1, IDH1, IDH2, or KIT at 1 of 111 treatment sites from 2013 through 2019. Clinical data were collected, through May 2022, by the Center for International Blood and Marrow Transplant Research. Exposure Centralized DNA sequencing of banked pretransplant remission blood samples. Main Outcomes and Measures The primary outcomes were overall survival and relapse. Day of transplant was considered day 0. Hazard ratios were reported using Cox proportional hazards regression models. Results Of 1075 patients tested, 822 had FLT3 internal tandem duplication (FLT3-ITD) and/or NPM1 mutated AML (median age, 57.1 years, 54% female). Among 371 patients in the discovery cohort, the persistence of NPM1 and/or FLT3-ITD variants in the blood of 64 patients (17.3%) in remission before undergoing transplant was associated with worse outcomes after transplant (2013-2017). Similarly, of the 451 patients in the validation cohort who had undergone transplant in 2018-2019, 78 patients (17.3%) with residual NPM1 and/or FLT3-ITD variants had higher rates of relapse at 3 years (68% vs 21%; difference, 47% [95% CI, 26% to 69%]; HR, 4.32 [95% CI, 2.98 to 6.26]; P < .001) and decreased survival at 3 years (39% vs 63%; difference, -24% [2-sided 95% CI, -39% to -9%]; HR, 2.43 [95% CI, 1.71 to 3.45]; P < .001). Conclusions and Relevance Among patients with acute myeloid leukemia in first remission prior to allogeneic hematopoietic cell transplant, the persistence of FLT3 internal tandem duplication or NPM1 variants in the blood at an allele fraction of 0.01% or higher was associated with increased relapse and worse survival compared with those without these variants. Further study is needed to determine whether routine DNA-sequencing testing for residual variants can improve outcomes for patients with acute myeloid leukemia.
Collapse
MESH Headings
- Female
- Humans
- Male
- Middle Aged
- Hematopoietic Stem Cell Transplantation
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Neoplasm, Residual/blood
- Neoplasm, Residual/diagnosis
- Neoplasm, Residual/genetics
- Nuclear Proteins/genetics
- Preoperative Care
- Retrospective Studies
- Sequence Analysis, DNA
- Recurrence
- Survival Analysis
Collapse
Affiliation(s)
- Laura W. Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Gege Gui
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Kristin M. Page
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
- Medical College of Wisconsin, Milwaukee
| | - Niveditha Ravindra
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Zoë C. Wong
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Georgia Andrew
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Devdeep Mukherjee
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Scott L. Zeger
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Stephen Spellman
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
- National Marrow Donor Program, Minneapolis, Minnesota
| | - Alan Howard
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
- National Marrow Donor Program, Minneapolis, Minnesota
| | - Karen Chen
- National Marrow Donor Program, Minneapolis, Minnesota
| | - Jeffery Auletta
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
- The Ohio State University College of Medicine, Columbus
| | - Steven M. Devine
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
| | | | | | | | | | - Wael Saber
- Medical College of Wisconsin, Milwaukee
- National Marrow Donor Program, Minneapolis, Minnesota
| | - Daniel J. Weisdorf
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
- University of Minnesota, Minneapolis
| | - Christopher S. Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Myeloid Malignancies Program, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
36
|
Tiong IS, Loo S. Targeting Measurable Residual Disease (MRD) in Acute Myeloid Leukemia (AML): Moving beyond Prognostication. Int J Mol Sci 2023; 24:4790. [PMID: 36902217 PMCID: PMC10003715 DOI: 10.3390/ijms24054790] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 03/06/2023] Open
Abstract
Measurable residual disease (MRD) assessment in acute myeloid leukemia (AML) has an established role in disease prognostication, particularly in guiding decisions for hematopoietic cell transplantation in first remission. Serial MRD assessment is now routinely recommended in the evaluation of treatment response and monitoring in AML by the European LeukemiaNet. The key question remains, however, if MRD in AML is clinically actionable or "does MRD merely portend fate"? With a series of new drug approvals since 2017, we now have more targeted and less toxic therapeutic options for the potential application of MRD-directed therapy. Recent approval of NPM1 MRD as a regulatory endpoint is also foreseen to drastically transform the clinical trial landscape such as biomarker-driven adaptive design. In this article, we will review (1) the emerging molecular MRD markers (such as non-DTA mutations, IDH1/2, and FLT3-ITD); (2) the impact of novel therapeutics on MRD endpoints; and (3) how MRD might be used as a predictive biomarker to guide therapy in AML beyond its prognostic role, which is the focus of two large collaborative trials: AMLM26 INTERCEPT (ACTRN12621000439842) and MyeloMATCH (NCT05564390).
Collapse
Affiliation(s)
- Ing S. Tiong
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- The Alfred Hospital, Melbourne, VIC 3004, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia
| | - Sun Loo
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- The Northern Hospital, Epping, VIC 3076, Australia
| |
Collapse
|
37
|
Krigstein M, Iland HJ, Wei AH. Applying molecular measurable residual disease testing in acute myeloid leukaemia. Pathology 2023; 55:1-7. [PMID: 36503638 DOI: 10.1016/j.pathol.2022.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
Molecular testing in acute myeloid leukaemia (AML) has continued to dramatically advance in recent years, facilitating the ability to detect residual disease at exponentially lower levels. With the advent of the recently updated ELN consensus recommendations, there is increasing complexity to ordering and interpreting measurable residual disease (MRD) assays in AML. We outline the technology itself in conjunction with the relevant testing timepoints, clinically significant thresholds and potential prognostic and therapeutic significance of MRD testing for the major molecular targets in AML. This practical overview should assist haematologists in incorporating molecular MRD assays routinely into their personalised AML clinical management.
Collapse
Affiliation(s)
- Michael Krigstein
- Department of Haematology, St Vincent's Hospital, Sydney, NSW, Australia.
| | - Harry J Iland
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Andrew H Wei
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Vic, Australia
| |
Collapse
|
38
|
Lee JM, Park S, Hwang I, Kang D, Cho BS, Kim HJ, Ahn A, Kim M, Kim Y. FLT3-ITD Measurable Residual Disease Monitoring in Acute Myeloid Leukemia Using Next-Generation Sequencing. Cancers (Basel) 2022; 14:6121. [PMID: 36551616 PMCID: PMC9776673 DOI: 10.3390/cancers14246121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
The in-frame internal tandem duplication (ITD) of the FMS-like tyrosine kinase 3 (FLT3) gene is an important negative prognostic marker in acute myeloid leukemia (AML). FLT3-ITD monitoring is essential for patients at relapse or those receiving FLT3-targeted therapies. Fragment analysis (FA) is commonly used to detect and quantify FLT3-ITDs; however, detecting low-burden FLT3-ITDs after a treatment is challenging. We, therefore, developed a customized, next-generation sequencing (NGS)-based FLT3-ITD assay that includes a new ITD-tracing algorithm, "SEED", optimized for measurable residual disease (MRD) monitoring. NGS-SEED showed an enhanced sensitivity (0.001%) and has a superior performance over conventional fragment analysis. We further investigated the prognostic impact of MRD analyzed by NGS-SEED in AML patients who underwent allogeneic hematopoietic stem cell transplantation (HSCT). Our assay showed that the MRD assessed before and after HSCT were significantly associated with a risk of relapse and a poor overall survival, respectively, in a time-dependent analysis. Thus, this report highlighted the prognostic value of serial MRD monitoring using a sensitive method in a clinical setting of AML patients with FLT3-ITD.
Collapse
Affiliation(s)
- Jong-Mi Lee
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Silvia Park
- Department of Hematology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Insik Hwang
- Dow Biomedica Inc., Seoul 05771, Republic of Korea
| | - Dain Kang
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Byung Sik Cho
- Department of Hematology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hee-Je Kim
- Department of Hematology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Ari Ahn
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Myungshin Kim
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Yonggoo Kim
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|