51
|
Zhao Y, Guo H, Chang Y. MRD-directed and risk-adapted individualized stratified treatment of AML. Chin J Cancer Res 2023; 35:451-469. [PMID: 37969959 PMCID: PMC10643342 DOI: 10.21147/j.issn.1000-9604.2023.05.04] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023] Open
Abstract
Measurable residual disease (MRD) has been widely recognized as a biomarker for deeply evaluating complete remission (CR), predicting relapse, guiding pre-emptive interventions, and serving as an endpoint surrogate for drug testing. However, despite the emergence of new technologies, there remains a lack of comprehensive understanding regarding the proper techniques, sample materials, and optimal time points for MRD assessment. In this review, we summarized the MRD methods, sample sources, and evaluation frequency according to the risk category of the European Leukemia Net (ELN) 2022. Additionally, we emphasize the importance of properly utilizing and combining these technologies. We have also refined the flowchart outlining each time point for pre-emptive interventions and intervention paths. The evaluation of MRD in acute myeloid leukemia (AML) is sophisticated, clinically applicable, and technology-dependent, and necessitates standardized approaches and further research.
Collapse
Affiliation(s)
- Yijing Zhao
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Hanfei Guo
- Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto 94304, USA
- the First Hospital of Jilin University, Cancer Center, Changchun 133021, China
| | - Yingjun Chang
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| |
Collapse
|
52
|
Núñez-Torrón Stock C, Jiménez Chillón C, Martín Moro F, Marquet Palomanes J, Velázquez Kennedy K, Piris Villaespesa M, Roldán Santiago E, Rodríguez Martín E, Chinea Rodríguez A, García Gutiérrez V, Moreno Jiménez G, López Jiménez J, Herrera Puente P. Patients with secondary acute myeloid leukemia undergoing allogeneic stem-cell transplant have inferior outcomes than de novo acute myeloid leukemia regardless minimal residual disease level by flow cytometry. Hematol Oncol 2023; 41:753-761. [PMID: 37081742 DOI: 10.1002/hon.3160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 03/03/2023] [Accepted: 04/12/2023] [Indexed: 04/22/2023]
Abstract
Secondary acute myeloid leukemia (s-AML) patients have a poor prognosis and currently the only curative therapy is allogeneic stem-cell transplant (HSCT). However, we do not yet know whether transplantation is sufficient to reverse the poor prognosis compared to de novo AML patients. We analyzed survival after HSCT comparing a cohort of 58 patients with s-AML versus 52 de novo patients who were transplanted between 2012 and 2020. Patients with s-AML had worse event-free survival (EFS) (p = 0.001) and overall survival (OS) (p < 0.001) compared to de novo AML due to an increased risk of relapse (p = 0.06) and non-relapse mortality (p = 0.03). The main difference in survival was observed in patients who achieved complete remission (CR) before HSCT (EFS p = 0.002 OS and <0.001), regardless minimal residual disease (MRD) by |multiparametric flow cytometry cohorts. In patients transplanted with active disease (AD), the prognosis was adverse in both s-AML and de novo AML groups (EFS p = 0.869 and OS p = 0.930). After excluding patients with AD, we stratified the cohort according to conditioning intensity, noticing that s-AML who received MAC had comparable outcomes to de novo AML, but the survival differences remained among reduce intensity conditioning group. In conclusion, transplanted s-AML patients have worse survival among patients in CR before HSCT, regardless of MRD level by flow cytometry compared to de novo AML. MAC patients had similar outcomes irrespective of leukemia ontogeny.
Collapse
Affiliation(s)
- Claudia Núñez-Torrón Stock
- Departamento de Hematología y Hemoterapia, Hospital Universitario Infanta Sofía, Madrid, Spain
- Universidad Alcalá de Henares, Alcala de Henares, Spain
- Universidad Europea de Madrid, Madrid, Spain
| | - Carlos Jiménez Chillón
- Departamento de Hematología y Hemoterapia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Fernando Martín Moro
- Departamento de Hematología y Hemoterapia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Juan Marquet Palomanes
- Departamento de Hematología y Hemoterapia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Kyra Velázquez Kennedy
- Departamento de Hematología y Hemoterapia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | | | | | | | - Valentín García Gutiérrez
- Universidad Alcalá de Henares, Alcala de Henares, Spain
- Departamento de Hematología y Hemoterapia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Gemma Moreno Jiménez
- Departamento de Hematología y Hemoterapia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier López Jiménez
- Universidad Alcalá de Henares, Alcala de Henares, Spain
- Departamento de Hematología y Hemoterapia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Pilar Herrera Puente
- Universidad Alcalá de Henares, Alcala de Henares, Spain
- Departamento de Hematología y Hemoterapia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| |
Collapse
|
53
|
Jaramillo S, Le Cornet L, Kratzmann M, Krisam J, Görner M, Hänel M, Röllig C, Wass M, Scholl S, Ringhoffer M, Reichart A, Steffen B, Kayser S, Mikesch JH, Schaefer-Eckart K, Schubert J, Geer T, Martin S, Kieser M, Sauer T, Kriegsmann K, Hundemer M, Serve H, Bornhäuser M, Müller-Tidow C, Schlenk RF. Q-HAM: a multicenter upfront randomized phase II trial of quizartinib and high-dose Ara-C plus mitoxantrone in relapsed/refractory AML with FLT3-ITD. Trials 2023; 24:591. [PMID: 37715270 PMCID: PMC10504729 DOI: 10.1186/s13063-023-07421-x] [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: 11/26/2021] [Accepted: 05/27/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND About 50% of older patients with acute myeloid leukemia (AML) fail to attain complete remission (CR) following cytarabine plus anthracycline-based induction therapy. Salvage chemotherapy regimens are based on high-dose cytarabine (HiDAC), which is frequently combined with mitoxantrone (HAM regimen). However, CR rates remain low, with less than one-third of the patients achieving a CR. FLT3-ITD has consistently been identified as an unfavorable molecular marker in both relapsed and refractory (r/r)-AML. One-quarter of patients who received midostaurin are refractory to induction therapy and relapse rate at 2 years exceeds 40%. The oral second-generation bis-aryl urea tyrosine kinase inhibitor quizartinib is a very selective FLT3 inhibitor, has a high capacity for sustained FLT3 inhibition, and has an acceptable toxicity profile. METHODS In this multicenter, upfront randomized phase II trial, all patients receive quizartinib combined with HAM (cytarabine 3g/m2 bidaily day one to day three, mitoxantrone 10mg/m2 days two and three) during salvage therapy. Efficacy is assessed by comparison to historical controls based on the matched threshold crossing approach with achievement of CR, complete remission with incomplete hematologic recovery (CRi), or complete remission with partial recovery of peripheral blood counts (CRh) as primary endpoint. During consolidation therapy (chemotherapy and allogeneic hematopoietic cell transplantation), patients receive either prophylactic quizartinib therapy or measurable residual disease (MRD)-triggered preemptive continuation therapy with quizartinib according to up-front randomization. The matched threshold crossing approach is a novel study-design to enhance the classic single-arm trial design by including matched historical controls from previous clinical studies. It overcomes common disadvantages of single-armed and small randomized studies, since the expected outcome of the observed study population can be adjusted based on the matched controls with a comparable distribution of known prognostic and predictive factors. Furthermore, balanced treatment groups lead to stable statistical models. However, one of the limitations of our study is the inability to adjust for unobserved or unknown confounders. Addressing the primary endpoint, CR/CRi/CRh after salvage therapy, the maximal sample size of 80 patients is assessed generating a desirable power of the used adaptive design, assuming a logistic regression is performed at a one-sided significance level α=0.05, the aspired power is 0.8, and the number of matching partners per intervention patient is at least 1. After enrolling 20 patients, the trial sample size will be recalculated in an interim analysis based on a conditional power argument. CONCLUSION Currently, there is no commonly accepted standard for salvage chemotherapy treatment. The objective of the salvage therapy is to reduce leukemic burden, achieve the best possible remission, and perform a hemopoietic stem-cell transplantation. Thus, in patients with FLT3-ITD mutation, the comparison of quizartinib with intensive salvage therapy versus chemotherapy alone appears as a logical consequence in terms of efficacy and safety. ETHICS AND DISSEMINATION Ethical approval and approvals from the local and federal competent authorities were granted. Trial results will be reported via peer-reviewed journals and presented at conferences and scientific meetings. TRIAL REGISTRATION ClinicalTrials.gov NCT03989713; EudraCT Number: 2018-002675-17.
Collapse
Affiliation(s)
- Sonia Jaramillo
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.
| | - Lucian Le Cornet
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Markus Kratzmann
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Johannes Krisam
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Martin Görner
- Department of Hematology, Oncology and Palliative Medicine, Community Hospital Bielefeld, Bielefeld, Germany
| | - Mathias Hänel
- Department of Medicine III, Hospital Chemnitz gGmbH, Chemnitz, Germany
| | - Christoph Röllig
- Department of Medicine and Polyclinic I, TU Dresden University Hospital, Dresden, Germany
| | - Maxi Wass
- Department of Medicine IV, Halle (Saale) University Hospital, Halle, Germany
| | - Sebastian Scholl
- Department of Medicine II, Jena University Hospital, Jena, Germany
| | - Mark Ringhoffer
- Department of Medicine, III, Hospital Karlsruhe, Karlsruhe, Germany
| | - Alexander Reichart
- Department of Hematology, Oncology and Palliative Medicine, Hospital Winnenden, Winnenden, Germany
| | - Björn Steffen
- Department of Medicine II, Frankfurt University Hospital, Frankfurt, Germany
| | - Sabine Kayser
- Department of Medicine I - Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | | | | | - Jörg Schubert
- Department of Inner Medicine II, Elbland Hospital Riesa, Riesa, Germany
| | - Thomas Geer
- Department of Medicine II, Diaconal Hospital Schwäbisch-Hall, Schwäbisch Hall, Germany
| | - Sonja Martin
- Department of Hematology, Oncology and Palliative Medicine, Robert-Bosch Hospital, Stuttgart, Germany
| | - Meinhard Kieser
- Department of Hematology, Oncology and Palliative Medicine, Robert-Bosch Hospital, Stuttgart, Germany
| | - Tim Sauer
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina Kriegsmann
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Hundemer
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hubert Serve
- Department of Medicine II, Frankfurt University Hospital, Frankfurt, Germany
| | - Martin Bornhäuser
- Department of Medicine and Polyclinic I, TU Dresden University Hospital, Dresden, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard F Schlenk
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
54
|
Ravindra N, Dillon LW, Gui G, Smith M, Gondek LP, Jones RJ, Corner A, Hourigan CS, Ambinder AJ. Persistent IDH mutations are not associated with increased relapse or death in patients with IDH-mutated acute myeloid leukemia undergoing allogeneic hematopoietic cell transplant with post-transplant cyclophosphamide. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.14.23294087. [PMID: 37662423 PMCID: PMC10473775 DOI: 10.1101/2023.08.14.23294087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The presence of measurable residual disease (MRD) prior to an allogeneic hematopoietic transplant (alloHCT) in Acute Myeloid Leukemia (AML) has been shown to be associated with an increased risk of post-transplant relapse. Since the Isocitrate Dehydrogenase genes (IDH1/2) are mutated in a considerable proportion of patients with AML, we studied if these mutations would serve as useful targets for MRD. Fifty-five IDH-mutated AML patients undergoing non-myeloablative alloHCT with post-transplant cyclophosphamide at a single center were sequenced at baseline using a multi-gene panel followed by targeted testing for persistent IDH mutations at the pre- and post-alloHCT timepoints by digital droplet PCR or error-corrected next generation sequencing. The cohort included patients who had been treated with IDH inhibitors pre- and post-transplant (20% and 17% for IDH1 and 38% and 28% for IDH2). Overall, 55% of patients analyzed had detectable IDH mutations during complete remission prior to alloHCT. However, there were no statistically significant differences in overall survival (OS), relapse-free survival (RFS), and cumulative incidence of relapse (CIR) at 3 years between patients who tested positive or negative for a persistent IDH mutation during remission (OS: IDH1 p=1, IDH2 p=0.87; RFS: IDH1 p=0.71, IDH2 p= 0.78; CIR: IDH1 p=0.92, IDH2 p=0.97). There was also no difference in the prevalence of persistent IDH mutation between patients who did and did not receive an IDH inhibitor (p=0.59). Mutational profiling of available relapse samples showed that 8 out of 9 patients still exhibited the original IDH mutation, indicating that the IDH mutations remained stable through the course of the disease. This study demonstrates that persistent IDH mutations during remission is not associated with inferior clinical outcomes after alloHCT in patients with AML.
Collapse
Affiliation(s)
- Niveditha Ravindra
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Laura W. Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Gege Gui
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Matthew Smith
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Lukasz P. Gondek
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Richard J. Jones
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Adam Corner
- Bio-Rad Laboratories, Digital Biology Group, 5731, W. Las Positias Blvd, Pleasanton, CA
| | - 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
|
55
|
Sartor C, Brunetti L, Audisio E, Cignetti A, Zannoni L, Cristiano G, Nanni J, Ciruolo R, Zingarelli F, Ottaviani E, Patuelli A, Bandini L, Forte D, Sciabolacci S, Cardinali V, Papayannidis C, Cavo M, Martelli MP, Curti A. A venetoclax and azacitidine bridge-to-transplant strategy for NPM1-mutated acute myeloid leukaemia in molecular failure. Br J Haematol 2023; 202:599-607. [PMID: 37226312 DOI: 10.1111/bjh.18887] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023]
Abstract
NPM1-mutated acute myeloid leukaemia (NPM1mut AML) represents a mostly favourable/intermediate risk disease that benefits from allogeneic haematopoietic stem cell transplantation (HSCT) in case of measurable residual disease (MRD) relapse or persistence after induction chemotherapy. Although the negative prognostic role of pre-HSCT MRD is established, no recommendations are available for the management of peri-transplant molecular failure (MF). Based on the efficacy data of venetoclax (VEN)-based treatment in NPM1mut AML older patients, we retrospectively analysed the off-label combination of VEN plus azacitidine (AZA) as bridge-to-transplant strategy in 11 NPM1mut MRD-positive fit AML patients. Patients were in MRD-positive complete remission (CRMRDpos ) at the time of treatment: nine in molecular relapse and two in molecular persistence. After a median number of two cycles (range 1-4) of VEN-AZA, 9/11 (81.8%) achieved CRMRD -negative (CRMRDneg ). All 11 patients proceeded to HSCT. With a median follow-up from treatment start of 26 months, and a median post-HSCT follow-up of 19 months, 10/11 patients are alive (1 died from non-relapse mortality), and 9/10 patients are in MRDneg status. This patient series highlights the efficacy and safety of VEN-AZA to prevent overt relapse, achieve deep responses and preserve patient fitness before HSCT, in patients with NPM1mut AML in MF.
Collapse
Affiliation(s)
- C Sartor
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - L Brunetti
- Clinica di Ematologia, Azienda Ospedaliero-Universitaria Ospedali Riuniti delle Marche, Ancona, Italy
| | - E Audisio
- SC Ematologia, Dipartimento di Ematologia e Oncologia, AO Città della Salute e della Scienza di Torino, Turin, Italy
| | - A Cignetti
- Department of Hematology and Cell Therapy, A.O. Ordine Mauriziano, Turin, Italy
| | - L Zannoni
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - G Cristiano
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - J Nanni
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - R Ciruolo
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - F Zingarelli
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - E Ottaviani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli" Bologna, Bologna, Italy
| | - A Patuelli
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - L Bandini
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - D Forte
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
| | - S Sciabolacci
- Institute of Hematology, Centro Ricerche Emato-Oncologiche, Ospedale S. Maria della Misericordia, University of Perugia, Perugia, Italy
| | - V Cardinali
- Institute of Hematology, Centro Ricerche Emato-Oncologiche, Ospedale S. Maria della Misericordia, University of Perugia, Perugia, Italy
| | - C Papayannidis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli" Bologna, Bologna, Italy
| | - M Cavo
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Ematologia "Seràgnoli", Università degli Studi di Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli" Bologna, Bologna, Italy
| | - M P Martelli
- Institute of Hematology, Centro Ricerche Emato-Oncologiche, Ospedale S. Maria della Misericordia, University of Perugia, Perugia, Italy
| | - A Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli" Bologna, Bologna, Italy
| |
Collapse
|
56
|
Short NJ, Ong F, Ravandi F, Nogueras-Gonzalez G, Kadia TM, Daver N, DiNardo CD, Konopleva M, Borthakur G, Oran B, Al-Atrash G, Mehta R, Jabbour EJ, Yilmaz M, Issa GC, Maiti A, Champlin RE, Kantarjian H, Shpall EJ, Popat U. Impact of type of induction therapy on outcomes in older adults with AML after allogeneic stem cell transplantation. Blood Adv 2023; 7:3573-3581. [PMID: 37104058 PMCID: PMC10368841 DOI: 10.1182/bloodadvances.2022009632] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Although venetoclax-based lower-intensity regimens have greatly improved outcomes for older adults with acute myeloid leukemia (AML) who are unfit for intensive chemotherapy, the optimal induction for older patients with newly diagnosed AML who are suitable candidates for hematopoietic stem cell transplant (HSCT) is controversial. We retrospectively analyzed the post HSCT outcomes of 127 patients ≥60 years of age who received induction therapy at our institution with intensive chemotherapy (IC; n = 44), lower-intensity therapy (LIT) without venetoclax (n = 29), or LIT with venetoclax (n = 54) and who underwent allogeneic HSCT in the first remission. The 2-year relapse-free survival (RFS) was 60% with LIT with venetoclax vs 54% with IC, and 41% with LIT without venetoclax; the 2-year overall survival (OS) was 72% LIT with venetoclax vs 58% with IC, and 41% with LIT without venetoclax. The benefit of LIT with venetoclax induction was greatest in patients with adverse-risk AML (2-year OS: 74%, 46%, and 29%, respectively). Induction with LIT, with or without venetoclax, was associated with the lowest rate of nonrelapse mortality (NRM) (2-year NRM: 17% vs 27% with IC; P = .04). Using multivariate analysis, the type of induction therapy did not significantly affect any of the post HSCT outcomes evaluated; hematopoietic cell transplantation-specific comorbidity index was the only factor that independently predicted RFS and OS. LIT plus venetoclax followed by HSCT is a feasible treatment strategy in older, fit, HSCT-eligible patients with newly diagnosed AML and may be particularly beneficial for those with adverse-risk disease.
Collapse
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Faustine Ong
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Tapan M. Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Betul Oran
- Department of Stem Cell Transplantation & Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation & Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rohtesh Mehta
- Department of Stem Cell Transplantation & Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elias J. Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard E Champlin
- Department of Stem Cell Transplantation & Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation & Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Uday Popat
- Department of Stem Cell Transplantation & Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
57
|
Boscaro E, Urbino I, Catania FM, Arrigo G, Secreto C, Olivi M, D'Ardia S, Frairia C, Giai V, Freilone R, Ferrero D, Audisio E, Cerrano M. Modern Risk Stratification of Acute Myeloid Leukemia in 2023: Integrating Established and Emerging Prognostic Factors. Cancers (Basel) 2023; 15:3512. [PMID: 37444622 DOI: 10.3390/cancers15133512] [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: 06/06/2023] [Revised: 07/02/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
An accurate estimation of AML prognosis is complex since it depends on patient-related factors, AML manifestations at diagnosis, and disease genetics. Furthermore, the depth of response, evaluated using the level of MRD, has been established as a strong prognostic factor in several AML subgroups. In recent years, this rapidly evolving field has made the prognostic evaluation of AML more challenging. Traditional prognostic factors, established in cohorts of patients treated with standard intensive chemotherapy, are becoming less accurate as new effective therapies are emerging. The widespread availability of next-generation sequencing platforms has improved our knowledge of AML biology and, consequently, the recent ELN 2022 recommendations significantly expanded the role of new gene mutations. However, the impact of rare co-mutational patterns remains to be fully disclosed, and large international consortia such as the HARMONY project will hopefully be instrumental to this aim. Moreover, accumulating evidence suggests that clonal architecture plays a significant prognostic role. The integration of clinical, cytogenetic, and molecular factors is essential, but hierarchical methods are reaching their limit. Thus, innovative approaches are being extensively explored, including those based on "knowledge banks". Indeed, more robust prognostic estimations can be obtained by matching each patient's genomic and clinical data with the ones derived from very large cohorts, but further improvements are needed.
Collapse
Affiliation(s)
- Eleonora Boscaro
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Irene Urbino
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Federica Maria Catania
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Giulia Arrigo
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Carolina Secreto
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Matteo Olivi
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Stefano D'Ardia
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Chiara Frairia
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Valentina Giai
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Roberto Freilone
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Dario Ferrero
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy
| | - Ernesta Audisio
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Marco Cerrano
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| |
Collapse
|
58
|
Abou Dalle I, Labopin M, Kröger N, Schroeder T, Finke J, Stelljes M, Neubauer A, Blaise D, Yakoub-Agha I, Salmenniemi U, Forcade E, Itäla-Remes M, Dreger P, Bug G, Passweg J, Heuser M, Choi G, Brissot E, Giebel S, Nagler A, Ciceri F, Bazarbachi A, Mohty M. Impact of disease burden on clinical outcomes of AML patients receiving allogeneic hematopoietic cell transplantation: a study from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Bone Marrow Transplant 2023; 58:784-790. [PMID: 37041215 DOI: 10.1038/s41409-023-01961-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/27/2023] [Accepted: 03/16/2023] [Indexed: 04/13/2023]
Abstract
Pre-transplant detectable measurable residual disease (MRD) is still associated with high risk of relapse and poor outcomes in acute myeloid leukemia (AML). We aimed at evaluating the impact of disease burden on prediction of relapse and survival in patients receiving allogeneic hematopoietic cell transplantation (allo-HCT) in first remission (CR1). We identified a total of 3202 adult AML patients, of these 1776 patients were in CR1 and MRD positive and 1426 patients were primary refractory at time of transplant. After a median follow-up of 24.4 months, non-relapse mortality and relapse rate were significantly higher in the primary refractory group compared to the CR1 MRD positive group (Hazards Ratio (HR) = 1.82 (95% CI: 1.47-2.24) p < 0.001 and HR = 1.54 (95% CI: 1.34-1.77), p < 0.001), respectively. Leukemia-free survival (LFS) and overall survival (OS) were significantly worse in the primary refractory group (HR = 1.61 (95% CI: 1.44-1.81), p < 0.001 and HR = 1.71 (95% CI: 1.51-1.94), p < 0.001, respectively). Our real-life data suggest that patients in CR1 and MRD positive at time of transplant could still be salvaged by allo-HCT with a 2-year OS of 63%, if negative MRD cannot be obtained and their outcomes are significantly better than patients transplanted with active disease.
Collapse
Affiliation(s)
- Iman Abou Dalle
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Myriam Labopin
- Department of Haematology, Hopital Saint-Antoine, INSERM, Paris, France
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Schroeder
- Dept. of Bone Marrow Transplantation, University Hospital, Essen, Germany
| | - Jürgen Finke
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Matthias Stelljes
- Department of Medicine A/Hematology and Oncology, University of Muenster, Muenster, Germany
| | - Andreas Neubauer
- Klinik für Innere Medizin mit SP Hämatologie, Onkologie und Immunologie, Philipps-Universität, Marburg, Germany
| | | | | | - Urpu Salmenniemi
- Department of Hematology, Helsinki University Hospital, Comprehensive Cancer Center and University of Helsinki, Helsinki, Finland
| | - Edouard Forcade
- CHU Bordeaux, Service d'hématologie Clinique et Thérapie Cellulaire, 33000, Pessac, France
| | - Maija Itäla-Remes
- Department of Medicine, Turku University Central Hospital, PL 52, Turku, Finland
| | - Peter Dreger
- Department Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Gesine Bug
- Department of Medicine II, University Hospital, Frankfurt, Germany
| | - Jakob Passweg
- EBMT Activity Survey Office, Hematology Division, University Hospital, Basel, Switzerland
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Goda Choi
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eolia Brissot
- Department of Hematology, Hospital Saint Antoine, Paris, France
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie Institute, Oncology Center-Gliwice, Gliwice, Poland
| | - Arnon Nagler
- Department of Bone Marrow Transplantation, Chaim Sheba Medical Center-Tel-Hashomer, Tel-Hashomer, Israel
| | - Fabio Ciceri
- Ospedale San Raffaele s.r.l., Haematology and BMT, Milano, Italy
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Mohamad Mohty
- Department of Haematology, Hopital Saint-Antoine, INSERM, Paris, France
| |
Collapse
|
59
|
El Chaer F, Hourigan CS, Zeidan AM. How I treat AML incorporating the updated classifications and guidelines. Blood 2023; 141:2813-2823. [PMID: 36758209 PMCID: PMC10447497 DOI: 10.1182/blood.2022017808] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/23/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
The European LeukemiaNet recently revised both the clinical (2022) and measurable residual disease testing (2021) guidelines for acute myeloid leukemia (AML). The updated World Health Organization and International Consensus Classification for myeloid neoplasms were also published in 2022. Together, these documents update the classification, risk stratification, prognostication, monitoring recommendations, and response assessment of patients with AML. Increased appreciation of the genetic drivers of AML over the past decade and our increasingly sophisticated understanding of AML biology have been translated into novel therapies and more complex clinical treatment guidelines. Somatic genetic abnormalities and germ line predispositions now define and guide treatment and counseling for the subtypes of this hematologic malignancy. In this How I Treat article, we discuss how we approach AML in daily clinical practice, considering the recent updates in the context of new treatments and discoveries over the past decade.
Collapse
Affiliation(s)
- Firas El Chaer
- Division of Hematology and Oncology, Department of Medicine, University of Virginia, Charlottesville, VA
| | - 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
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine and Yale Comprehensive Cancer Center, Yale University, New Haven, CT
| |
Collapse
|
60
|
Pitts HA, Cheng CK, Cheung JS, Sun MKH, Yung YL, Chan HY, Wong RSM, Yip SF, Lau KN, Wong WS, Raghupathy R, Chan NPH, Ng MHL. SPINK2 Protein Expression Is an Independent Adverse Prognostic Marker in AML and Is Potentially Implicated in the Regulation of Ferroptosis and Immune Response. Int J Mol Sci 2023; 24:ijms24119696. [PMID: 37298647 DOI: 10.3390/ijms24119696] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
There is an urgent need for the identification as well as clinicopathological and functional characterization of potent prognostic biomarkers and therapeutic targets in acute myeloid leukemia (AML). Using immunohistochemistry and next-generation sequencing, we investigated the protein expression as well as clinicopathological and prognostic associations of serine protease inhibitor Kazal type 2 (SPINK2) in AML and examined its potential biological functions. High SPINK2 protein expression was an independent adverse biomarker for survival and an indicator of elevated therapy resistance and relapse risk. SPINK2 expression was associated with AML with an NPM1 mutation and an intermediate risk by cytogenetics and European LeukemiaNet (ELN) 2022 criteria. Furthermore, SPINK2 expression could refine the ELN2022prognostic stratification. Functionally, an RNA sequencing analysis uncovered a potential link of SPINK2 with ferroptosis and immune response. SPINK2 regulated the expression of certain P53 targets and ferroptosis-related genes, including SLC7A11 and STEAP3, and affected cystine uptake, intracellular iron levels and sensitivity to erastin, a specific ferroptosis inducer. Furthermore, SPINK2 inhibition consistently increased the expression of ALCAM, an immune response enhancer and promoter of T-cell activity. Additionally, we identified a potential small-molecule inhibitor of SPINK2, which requires further characterization. In summary, high SPINK2 protein expression was a potent adverse prognostic marker in AML and might represent a druggable target.
Collapse
Affiliation(s)
- Herbert Augustus Pitts
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi-Keung Cheng
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joyce Sin Cheung
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Murphy Ka-Hei Sun
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuk-Lin Yung
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hoi-Yun Chan
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Raymond S M Wong
- Sir Y.K. Pao Centre for Cancer, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sze-Fai Yip
- Department of Clinical Pathology, Tuen Mun Hospital, Hong Kong SAR, China
| | - Ka-Ngai Lau
- Department of Clinical Pathology, Tuen Mun Hospital, Hong Kong SAR, China
| | - Wai Shan Wong
- Pathology Department, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Radha Raghupathy
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Natalie P H Chan
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Margaret H L Ng
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
61
|
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: 1.0] [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
|
62
|
Guijarro F, Garrote M, Villamor N, Colomer D, Esteve J, López-Guerra M. Novel Tools for Diagnosis and Monitoring of AML. Curr Oncol 2023; 30:5201-5213. [PMID: 37366878 DOI: 10.3390/curroncol30060395] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/11/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
In recent years, major advances in the understanding of acute myeloid leukemia (AML) pathogenesis, together with technological progress, have led us into a new era in the diagnosis and follow-up of patients with AML. A combination of immunophenotyping, cytogenetic and molecular studies are required for AML diagnosis, including the use of next-generation sequencing (NGS) gene panels to screen all genetic alterations with diagnostic, prognostic and/or therapeutic value. Regarding AML monitoring, multiparametric flow cytometry and quantitative PCR/RT-PCR are currently the most implemented methodologies for measurable residual disease (MRD) evaluation. Given the limitations of these techniques, there is an urgent need to incorporate new tools for MRD monitoring, such as NGS and digital PCR. This review aims to provide an overview of the different technologies used for AML diagnosis and MRD monitoring and to highlight the limitations and challenges of current versus emerging tools.
Collapse
Affiliation(s)
- Francesca Guijarro
- Hematopathology Section, Pathology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Marta Garrote
- Hematopathology Section, Pathology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Neus Villamor
- Hematopathology Section, Pathology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Dolors Colomer
- Hematopathology Section, Pathology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Jordi Esteve
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Hematology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
| | - Mónica López-Guerra
- Hematopathology Section, Pathology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
63
|
Stempel JM, Xie Z, Bewersdorf JP, Stahl M, Zeidan AM. Evolution of Therapeutic Benefit Measurement Criteria in Myelodysplastic Syndromes/Neoplasms. Cancer J 2023; 29:203-211. [PMID: 37195777 DOI: 10.1097/ppo.0000000000000666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
ABSTRACT Myelodysplastic syndromes/neoplasms (MDS) are heterogeneous, clonal myeloid neoplasms characterized by ineffective hematopoiesis, progressive cytopenias, and an increased risk of progression to acute myeloid leukemia. The diversity in disease severity, morphology, and genetic landscape challenges not only novel drug development but also therapeutic response assessment. The MDS International Working Group (IWG) response criteria were first published in the year 2000 focusing on measures of blast burden reduction and hematologic recovery. Despite revision of the IWG criteria in 2006, correlation between IWG-defined responses and patient-focused outcomes, including long-term benefits, remains limited and has potentially contributed to failures of several phase III clinical trials. Several IWG 2006 criteria also lacked clear definitions leading to problems in practical applications and interobserver and intraobserver consistency of response reporting. Although the 2018 revision addressed lower-risk MDS, the most recent update in 2023 redefined responses for higher-risk MDS and has set out to provide clear definitions to enhance consistency while focusing on clinically meaningful outcomes and patient-centered responses. In this review, we analyze the evolution of the MDS response criteria, limitations, and areas of improvement.
Collapse
Affiliation(s)
- Jessica M Stempel
- From the Department of Internal Medicine, Hematology Section, Yale School of Medicine, New Haven, CT
| | - Zhuoer Xie
- Department of Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Harvard University, Boston, MA
| | - Amer M Zeidan
- From the Department of Internal Medicine, Hematology Section, Yale School of Medicine, New Haven, CT
| |
Collapse
|
64
|
Boyiadzis M, Zhang MJ, Chen K, Abdel-Azim H, Abid MB, Aljurf M, Bacher U, Badar T, Badawy SM, Battiwalla M, Bejanyan N, Bhatt VR, Brown VI, Castillo P, Cerny J, Copelan EA, Craddock C, Dholaria B, Perez MAD, Ebens CL, Gale RP, Ganguly S, Gowda L, Grunwald MR, Hashmi S, Hildebrandt GC, Iqbal M, Jamy O, Kharfan-Dabaja MA, Khera N, Lazarus HM, Lin R, Modi D, Nathan S, Nishihori T, Patel SS, Pawarode A, Saber W, Sharma A, Solh M, Wagner JL, Wang T, Williams KM, Winestone LE, Wirk B, Zeidan A, Hourigan CS, Litzow M, Kebriaei P, de Lima M, Page K, Weisdorf DJ. Impact of pre-transplant induction and consolidation cycles on AML allogeneic transplant outcomes: a CIBMTR analysis in 3113 AML patients. Leukemia 2023; 37:1006-1017. [PMID: 36310182 PMCID: PMC10148918 DOI: 10.1038/s41375-022-01738-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/06/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022]
Abstract
We investigated the impact of the number of induction/consolidation cycles on outcomes of 3113 adult AML patients who received allogeneic hematopoietic cell transplantation (allo-HCT) between 2008 and 2019. Patients received allo-HCT using myeloablative (MAC) or reduced-intensity (RIC) conditioning in first complete remission (CR) or with primary induction failure (PIF). Patients who received MAC allo-HCT in CR after 1 induction cycle had 1.3-fold better overall survival (OS) than 2 cycles to CR and 1.47-fold better than ≥3 cycles. OS after CR in 2 or ≥3 cycles was similar. Relapse risk was 1.65-fold greater in patients receiving ≥3 cycles to achieve CR. After RIC allo-HCT, the number of induction cycles to CR did not affect OS. Compared to CR in 1 cycle, relapse risk was 1.24-1.41-fold greater in patients receiving 2 or ≥3 cycles. For patients receiving only 1 cycle to CR, consolidation therapy prior to MAC allo-HCT was associated with improved OS vs. no consolidation therapy. Detectable MRD at the time of MAC allo-HCT did not impact outcomes while detectable MRD preceding RIC allo-HCT was associated with an increased risk of relapse. For allo-HCT in PIF, OS was significantly worse than allo-HCT in CR after 1-3 cycles.
Collapse
Affiliation(s)
| | - Mei-Jie Zhang
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Karen Chen
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hisham Abdel-Azim
- Loma Linda University School of Medicine, Cancer Center, Children Hospital and Medical Center, Loma Linda, CA, USA
| | - Muhammad Bilal Abid
- Divisions of Hematology/Oncology & Infectious Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mahmoud Aljurf
- Department of Oncology, King Faisal Specialist Hospital Center & Research, Riyadh, Saudi Arabia
| | - Ulrike Bacher
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Talha Badar
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Sherif M Badawy
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Nelli Bejanyan
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, FL, USA
| | - Vijaya Raj Bhatt
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Valerie I Brown
- Division of Pediatric Oncology/Hematology, Department of Pediatrics, Penn State Hershey Children's Hospital and College of Medicine, Hershey, PA, USA
| | - Paul Castillo
- UF Health Shands Children's Hospital, Gainesville, FL, USA
| | - Jan Cerny
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical Center, Worcester, MA, USA
| | - Edward A Copelan
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | | | | | - Miguel Angel Diaz Perez
- Department of Hematology/Oncology, Hospital Infantil Universitario Niño Jesus, Madrid, Spain
| | - Christen L Ebens
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Peter Gale
- Haematology Centre, Department of Immunology and Inflammation, Imperial College London, London, UK
| | | | - Lohith Gowda
- Yale Cancer Center and Yale School of Medicine, New Haven, CT, USA
| | - Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Shahrukh Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, UAE
| | | | - Madiha Iqbal
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Omer Jamy
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Nandita Khera
- Department of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Hillard M Lazarus
- University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - Richard Lin
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dipenkumar Modi
- Division of Oncology, Karmanos Cancer Center/Wayne State University, Detroit, MI, USA
| | - Sunita Nathan
- Section of Bone Marrow Transplant and Cell Therapy, Rush University Medical Center, Chicago, IL, USA
| | - Taiga Nishihori
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, FL, USA
| | - Sagar S Patel
- Transplant and Cellular Therapy Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Attaphol Pawarode
- Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI, USA
| | - Wael Saber
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin, Milwaukee, WI, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, GA, USA
| | - John L Wagner
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Trent Wang
- Division of Transplantation and Cellular Therapy, University of Miami, Miami, FL, USA
| | | | - Lena E Winestone
- Division of Allergy, Immunology, and Blood & Marrow Transplant, University of California San Francisco Benioff Children's Hospitals, San Francisco, CA, USA
| | - Baldeep Wirk
- Bone Marrow Transplant Program, Penn State Cancer Institute, Hershey, PA, USA
| | - Amer Zeidan
- Bridgeport Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark Litzow
- Division of Hematology and Transplant Center, Mayo Clinic Rochester, Rochester, MN, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Kristin Page
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Daniel J Weisdorf
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
65
|
Walter RB, Sandmaier BM, Othus M, Orvain C, Rodríguez-Arbolí E, Oshima MU, Schoch G, Davis C, Joachim Deeg H, Storb R. Comparison of reduced intensity and nonmyeloablative conditioning for adults with acute myeloid leukemia undergoing allogeneic hematopoietic cell transplantation in first or second remission. Bone Marrow Transplant 2023; 58:377-385. [PMID: 36577856 PMCID: PMC10170527 DOI: 10.1038/s41409-022-01909-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022]
Abstract
Reduced intensity conditioning (RIC) and nonmyeloablative (NMA) conditioning regimens have expanded use of allogeneic hematopoietic cell transplantation (HCT) in AML to include older and medically less-fit patients, but relative efficacies and toxicities remain poorly defined. Here, we analyzed outcomes from 343 adults transplanted in remission after RIC (n = 137) or NMA (n = 206) conditioning between 2006 and 2021. The characteristics of RIC and NMA HCT patients were similar except that RIC patients were younger and their time between most recent remission achievement and allografting was shorter. There were no significant differences in relapse risk, relapse-free survival (RFS), overall survival (OS), and non-relapse mortality (NRM) between RIC and NMA HCT patients, both overall (relapse: hazard ratio [HR] = 0.80, P = 0.27; RFS: HR = 0.93, P = 0.61; OS: HR = 0.93, P = 0.66; NRM: HR = 1.13, P = 0.59) and when patients were stratified by pre-HCT measurable residual disease (MRD) status. After multivariable adjustment, there was no statistically significant association between conditioning intensity and relapse (HR = 0.69, P = 0.088), RFS (HR = 0.86, P = 0.37), OS (HR = 0.89, P = 0.49), or NRM (HR = 1.37, P = 0.19). In this non-randomized cohort of adults undergoing allografting for AML in first or second remission at our center, we could not detect statistically significant differences in outcomes between those assigned to RIC and those assigned to NMA conditioning.
Collapse
Affiliation(s)
- Roland B Walter
- Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA.
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| | - Brenda M Sandmaier
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Megan Othus
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Corentin Orvain
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| | | | - Masumi U Oshima
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Gary Schoch
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chris Davis
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - H Joachim Deeg
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Rainer Storb
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
66
|
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: 82] [Impact Index Per Article: 41.0] [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
|
67
|
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: 4.0] [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
|
68
|
Tringale KR, Wolden SL, Karajannis M, Haque S, Pasquini L, Yildirim O, Rosenblum M, Benhamida JK, Cheung NK, Souweidane M, Basu EM, Pandit-Taskar N, Zanzonico PB, Humm JL, Kramer K. Outcomes of intraventricular 131-I-omburtamab and external beam radiotherapy in patients with recurrent medulloblastoma and ependymoma. J Neurooncol 2023; 162:69-78. [PMID: 36853490 PMCID: PMC10050019 DOI: 10.1007/s11060-022-04235-w] [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: 11/27/2022] [Accepted: 12/30/2022] [Indexed: 03/01/2023]
Abstract
PURPOSE Intraventricular compartmental radioimmunotherapy (cRIT) with 131-I-omburtamab is a potential therapy for recurrent primary brain tumors that can seed the thecal space. These patients often previously received external beam radiotherapy (EBRT) to a portion or full craniospinal axis (CSI) as part of upfront therapy. Little is known regarding outcomes after re-irradiation as part of multimodality therapy including cRIT. This study evaluates predictors of response, patterns of failure, and radiologic events after cRIT. METHODS Patients with recurrent medulloblastoma or ependymoma who received 131-I-omburtamab on a prospective clinical trial were included. Extent of disease at cRIT initiation (no evidence of disease [NED] vs measurable disease [MD]) was assessed as associated with progression-free (PFS) and overall survival (OS) by Kaplan-Meier analysis. RESULTS All 27 patients (20 medulloblastoma, 7 ependymoma) had EBRT preceding cRIT: most (22, 81%) included CSI (median dose 2340 cGy, boost to 5400 cGy). Twelve (44%) also received EBRT at relapse as bridging to cRIT. There were no cases of radionecrosis. At cRIT initiation, 11 (55%) medulloblastoma and 3 (43%) ependymoma patients were NED, associated with improved PFS (p = 0.002) and OS (p = 0.048) in medulloblastoma. Most relapses were multifocal. With medium follow-up of 3.0 years (95% confidence interval, 1.8-7.4), 6 patients remain alive with NED. CONCLUSION For patients with medulloblastoma, remission at time of cRIT was associated with significantly improved survival outcomes. Relapses are often multifocal, particularly in the setting of measurable disease at cRIT initiation. EBRT is a promising tool to achieve NED status at cRIT initiation, with no cases of radiation necrosis.
Collapse
Affiliation(s)
- Kathryn R Tringale
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthias Karajannis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luca Pasquini
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Onur Yildirim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc Rosenblum
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jamal K Benhamida
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nai-Kong Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark Souweidane
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neeta Pandit-Taskar
- Department of Nuclear Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pat B Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John L Humm
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
69
|
NPM 1 Mutations in AML-The Landscape in 2023. Cancers (Basel) 2023; 15:cancers15041177. [PMID: 36831522 PMCID: PMC9954410 DOI: 10.3390/cancers15041177] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Acute myeloid leukemia (AML) represents 80% of acute leukemia in adults and is characterized by clonal expansion of hematopoietic stem cells secondary to genomic mutations, rendering a selective growth advantage to the mutant clones. NPM1mut is found in around 30% of AML and clinically presents with leukocytosis, high blast percentage and extramedullary involvement. Considered as a "gate-keeper" mutation, NPM1mut appears to be a "first hit" in the process of leukemogenesis and development of overt leukemia. Commonly associated with other mutations (e.g., FLT 3, DNMT3A, TET2, SF3B1), NPM1 mutation in AML has an important role in diagnosis, prognosis, treatment and post-treatment monitoring. Several novel therapies targeting NPM1 are being developed in various clinical phases with demonstration of efficacy. In this review, we summarize the pathophysiology of the NPM1 gene mutation in AML, clinical implications and the novel targeted therapies to date.
Collapse
|
70
|
Meddi E, Savi A, Moretti F, Mallegni F, Palmieri R, Paterno G, Buzzatti E, Del Principe MI, Buccisano F, Venditti A, Maurillo L. Measurable Residual Disease (MRD) as a Surrogate Efficacy-Response Biomarker in AML. Int J Mol Sci 2023; 24:ijms24043062. [PMID: 36834477 PMCID: PMC9967250 DOI: 10.3390/ijms24043062] [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: 12/30/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
In acute myeloid leukemia (AML) many patients experience relapse, despite the achievement of morphological complete remission; therefore, conventional morphologic criteria are currently considered inadequate for assessing the quality of the response after treatment. Quantification of measurable residual disease (MRD) has been established as a strong prognostic marker in AML and patients that test MRD negative have lower relapse rates and better survival than those who test positive. Different techniques, varying in their sensitivity and applicability to patients, are available for the measurement of MRD and their use as a guide for selecting the most optimal post-remission therapy is an area of active investigation. Although still controversial, MRD prognostic value promises to support drug development serving as a surrogate biomarker, potentially useful for accelerating the regulatory approval of new agents. In this review, we will critically examine the methods used to detect MRD and its potential role as a study endpoint.
Collapse
Affiliation(s)
- Elisa Meddi
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Arianna Savi
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Federico Moretti
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Flavia Mallegni
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Raffaele Palmieri
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | | | - Elisa Buzzatti
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | | | - Francesco Buccisano
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
- Correspondence:
| | - Luca Maurillo
- Hematology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
71
|
Measurable Residual Disease and Clonal Evolution in Acute Myeloid Leukemia from Diagnosis to Post-Transplant Follow-Up: The Role of Next-Generation Sequencing. Biomedicines 2023; 11:biomedicines11020359. [PMID: 36830896 PMCID: PMC9953407 DOI: 10.3390/biomedicines11020359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
It has now been ascertained that acute myeloid leukemias-as in most type of cancers-are mixtures of various subclones, evolving by acquiring additional somatic mutations over the course of the disease. The complexity of leukemia clone architecture and the phenotypic and/or genotypic drifts that can occur during treatment explain why more than 50% of patients-in hematological remission-could relapse. Moreover, the complexity and heterogeneity of clone architecture represent a hindrance for monitoring measurable residual disease, as not all minimal residual disease monitoring methods are able to detect genetic mutations arising during treatment. Unlike with chemotherapy, which imparts a relatively short duration of selective pressure on acute myeloid leukemia clonal architecture, the immunological effect related to allogeneic hematopoietic stem cell transplant is prolonged over time and must be overcome for relapse to occur. This means that not all molecular abnormalities detected after transplant always imply inevitable relapse. Therefore, transplant represents a critical setting where a measurable residual disease-based strategy, performed during post-transplant follow-up by highly sensitive methods such as next-generation sequencing, could optimize and improve treatment outcome. The purpose of our review is to provide an overview of the role of next-generation sequencing in monitoring both measurable residual disease and clonal evolution in acute myeloid leukemia patients during the entire course of the disease, with special focus on the transplant phase.
Collapse
|
72
|
Ramos Elbal E, Fuster JL, Campillo JA, Galera AM, Cortés MB, Llinares ME, Jiménez I, Plaza M, Banaclocha HM, Galián JA, Blanquer Blanquer M, Martínez Sánchez MV, Muro M, Minguela A. Measurable residual disease study through three different methods can anticipate relapse and guide pre-emptive therapy in childhood acute myeloid leukemia. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:1446-1454. [PMID: 36598635 DOI: 10.1007/s12094-022-03042-z] [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: 11/23/2022] [Accepted: 12/04/2022] [Indexed: 01/05/2023]
Abstract
PURPOSE Although outcomes of children with acute myeloid leukemia (AML) have improved over the last decades, around one-third of patients relapse. Measurable (or minimal) residual disease (MRD) monitoring may guide therapy adjustments or pre-emptive treatments before overt hematological relapse. METHODS In this study, we review 297 bone marrow samples from 20 real-life pediatric AML patients using three MRD monitoring methods: multiparametric flow cytometry (MFC), fluorescent in situ hybridization (FISH) and polymerase chain reaction (PCR). RESULTS Patients showed a 3-year overall survival of 73% and a 3-year event-free survival of 68%. Global relapse rate was of 25%. All relapses were preceded by the reappearance of MRD detection by: (1) MFC (p = 0.001), (2) PCR and/or FISH in patients with an identifiable chromosomal translocation (p = 0.03) and/or (3) one log increase of Wilms tumor gene 1 (WT1) expression in two consecutive samples (p = 0.02). The median times from MRD detection to relapse were 26, 111, and 140 days for MFC, specific PCR and FISH, and a one log increment of WT1, respectively. CONCLUSIONS MFC, FISH and PCR are complementary methods that can anticipate relapse of childhood AML by weeks to several months. However, in our series, pre-emptive therapies were not able to prevent disease progression. Therefore, more sensitive MRD monitoring methods that further anticipate relapse and more effective pre-emptive therapies are needed.
Collapse
Affiliation(s)
- Eduardo Ramos Elbal
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - José Luis Fuster
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - José Antonio Campillo
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Ana María Galera
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Mar Bermúdez Cortés
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - María Esther Llinares
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Irene Jiménez
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Mercedes Plaza
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Helios Martínez Banaclocha
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - José Antonio Galián
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Miguel Blanquer Blanquer
- Haematology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - María Victoria Martínez Sánchez
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Manuel Muro
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Alfredo Minguela
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain.
| |
Collapse
|
73
|
Meur GL, Plesa A, Larcher MV, Fossard G, Barraco F, Loron S, Balsat M, Ducastelle-Leprêtre S, Gilis L, Thomas X, Ghesquières H, Tigaud I, Hayette S, Huet S, Sujobert P, Renault M, Thérèse RM, Michallet M, Labussière-Wallet H, Heiblig M. Impact on Outcome of Minimal Residual Disease after Hematopoietic Stem Cell Transplantation with Fludarabine, Amsacrine, and Cytosine Arabinoside-Busulfan Conditioning: A Retrospective Monocentric Study. Transplant Cell Ther 2023; 29:38.e1-38.e9. [PMID: 36108977 DOI: 10.1016/j.jtct.2022.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/10/2022] [Accepted: 09/05/2022] [Indexed: 02/09/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) after conditioning with a sequential association of fludarabine, amsacrine, and cytosine arabinoside (FLAMSA) followed by a reduced-intensity conditioning regimen has emerged for patients with high-risk acute myeloid leukemia (AML), especially in refractory or relapsing patients. Here we aimed to address retrospectively the impact of pretransplantation minimal residual disease (MRD) by flow cytometry on the outcomes of high-risk AML patients who underwent allo-HSCT after sequential FLAMSA-busulfan (FLAMSA-Bu)-based conditioning regimens. We included 165 high-risk AML patients who underwent transplantation after FLAMSA-BU in this retrospective single-center "real life" study. All patients received in vivo T cell depletion with antithymocyte globulin (5 mg/kg). MRD detection was based on a leukemia-associated immunophenotype using the European LeukemiaNet recommendations, with a threshold of .1%. Univariate and multivariate analyses were performed using R version 4.1.1 (R Foundation for Statistical Computing, Vienna, Austria). With a median follow-up of 4.0 years post-transplantation, the median overall survival (OS) was 54.9 months. Overall, 41 patients (24.8%) relapsed post-transplantation, with a resulting cumulative incidence of relapse (CIR) of 26.7% at 2 years and 34.0% at 5 years. Detectable MRD preceding allo-HSCT and refractory status were associated with worse median OS and CIR rates compared with patients without detectable MRD; however, OS was not significantly different between pre-HSCT MRD-positive and refractory patients (median, .7 year versus 2.0 years; P = .3). Conversely, pre-HSCT MRD negativity was associated with a reduced 2-year CIR. Neither European LeukemiaNet risk stratification nor age had a significant influence on OS. In the multivariate analysis, only pre-HSCT MRD positivity and lower conditioning regimen intensity were significantly associated with a poorer OS. The cumulative incidence of extensive chronic graft-versus-host disease at 2 years was 26.15%. The estimated nonrelapse mortality (NRM) of the entire cohort at 2 years was 23.1%, with age and unrelated donor identified as risk factors for higher NRM. Our data ahow that FLAMSA-Bu conditioning did not reverse the pejorative effect of detectable pre-HSCT MRD, suggesting that such patients should be offered alternative strategies before HSCT to reach deeper remission.
Collapse
Affiliation(s)
- Grégoire Le Meur
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France; Université Claude Bernard, Lyon, France.
| | - Adriana Plesa
- Laboratory of Cytology and Immunology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Marie-Virginie Larcher
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Gaëlle Fossard
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Fiorenza Barraco
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Sandrine Loron
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Marie Balsat
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | | | - Lila Gilis
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Xavier Thomas
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Hervé Ghesquières
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France; Université Claude Bernard, Lyon, France
| | - Isabelle Tigaud
- Laboratory of Cytogenetics, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Sandrine Hayette
- Laboratory of Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Sarah Huet
- Laboratory of Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Pierre Sujobert
- Laboratory of Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Myriam Renault
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France
| | - Rubio Marie Thérèse
- Department of Hematology, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365, Biopole de l'Université del Lorraine, Vendoeuvre les Nancy, France
| | | | | | - Maël Heiblig
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre Bénite, France; Université Claude Bernard, Lyon, France.
| |
Collapse
|
74
|
Malagola M, Polverelli N, Beghin A, Bolda F, Comini M, Farina M, Morello E, Radici V, Accorsi Buttini E, Bernardi S, Re F, Leoni A, Bonometti D, Brugnoni D, Lanfranchi A, Russo D. Bone marrow CD34+ molecular chimerism as an early predictor of relapse after allogeneic stem cell transplantation in patients with acute myeloid leukemia. Front Oncol 2023; 13:1133418. [PMID: 36950550 PMCID: PMC10025489 DOI: 10.3389/fonc.2023.1133418] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Background Minimal residual disease (MRD) monitoring is an important tool to optimally address post-transplant management of acute myeloid leukemia (AML) patients. Methods We retrospectively analyzed the impact of bone marrow CD34+ molecular chimerism and WT1 on the outcome of a consecutive series of 168 AML patients submitted to allogeneic stem cell transplantation. Results The cumulative incidence of relapse (CIR) was significantly lower in patients with donor chimerism on CD34+ cells ≥ 97.5% and WT1 < 213 copies/ABL x 10^4 both at 1st month (p=0.008 and p<0.001) and at 3rd month (p<0.001 for both). By combining chimerism and WT1 at 3rd month, 13 patients with chimerism < 97.5% or WT1 > 213 showed intermediate prognosis. 12 of these patients fell in this category because of molecular chimerism < 97.5% at a time-point in which WT1 was < 213. Conclusions Our results confirm that lineage-specific molecular chimerism and WT1 after allo-SCT (1st and 3rd month) are useful MRD markers. When considered together at 3rd month, CD34+ molecular chimerism could represent an earlier predictor of relapse compared to WT1. Further studies are necessary to confirm this preliminary observation.
Collapse
Affiliation(s)
- Michele Malagola
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- *Correspondence: Michele Malagola,
| | - Nicola Polverelli
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandra Beghin
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Federica Bolda
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Marta Comini
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Mirko Farina
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Enrico Morello
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Vera Radici
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Eugenia Accorsi Buttini
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Simona Bernardi
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA) , “ASST-Spedali Civili” Hospital of Brescia, Brescia, Italy
| | - Federica Re
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA) , “ASST-Spedali Civili” Hospital of Brescia, Brescia, Italy
| | - Alessandro Leoni
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA) , “ASST-Spedali Civili” Hospital of Brescia, Brescia, Italy
| | - Davide Bonometti
- Department of Hematology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Duilio Brugnoni
- Department of Laboratory Diagnostics, ASST Spedali Civili, Brescia, Italy
| | - Arnalda Lanfranchi
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Domenico Russo
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
75
|
Achieving MRD negativity in AML: how important is this and how do we get there? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:9-14. [PMID: 36485093 PMCID: PMC9820122 DOI: 10.1182/hematology.2022000323] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple studies have demonstrated that patients with acute myeloid leukemia (AML) who have measurable residual disease (MRD) detected during or after treatment have higher relapse rates and worse survival than similar patients testing negative. Updated response criteria for AML reflect the understanding that achievement of complete remission (CR) with no detectable MRD using high-sensitivity tests represents a superior response over conventional cytomorphological CR alone. Potential use cases for AML MRD testing are diverse and include patient selection for clinical trials and therapeutic assignment, early relapse detection and intervention during sequential monitoring, and drug development, including deep quantification for antileukemia efficacy and as a surrogate endpoint for overall survival in regulatory approvals. Testing for AML MRD has not, however, been harmonized, and many technical and clinical questions remain. The implications of MRD test results for specific therapeutic combinations, molecular subsets, test types, treatment time points, sample types, and patient characteristics remain incompletely defined. No perfect AML MRD test or testing strategy currently exists, and the evidence basis for clinical recommendations in this rare disease is sparse but growing. It is unproven whether conversion of an MRD test result from positive to negative by additional therapeutic intervention improves relapse risk and survival. Several national- and international-level consortia have recently been initiated to advance the generation and collection of evidence to support the use of AML MRD testing in clinical practice, drug development, and regulatory approvals.
Collapse
|
76
|
Blachly JS, Walter RB, Hourigan CS. The present and future of measurable residual disease testing in acute myeloid leukemia. Haematologica 2022; 107:2810-2822. [PMID: 36453518 PMCID: PMC9713561 DOI: 10.3324/haematol.2022.282034] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Indexed: 12/04/2022] Open
Abstract
Considerable progress has been made in the past several years in the scientific understanding of, and available treatments for, acute myeloid leukemia (AML). Achievement of a conventional remission, evaluated cytomorphologically via small bone marrow samples, is a necessary but not sufficient step toward cure. It is increasingly appreciated that molecular or immunophenotypic methods to identify and quantify measurable residual disease (MRD) - populations of leukemia cells below the cytomorphological detection limit - provide refined information on the quality of response to treatment and prediction of the risk of AML recurrence and leukemia-related deaths. The principles and practices surrounding MRD remain incompletely determined however and the genetic and immunophenotypic heterogeneity of AML may prevent a one-sizefits- all approach. Here, we review the current approaches to MRD testing in AML, discuss strengths and limitations, highlight recent technological advances that may improve such testing, and summarize ongoing initiatives to generate the clinical evidence needed to advance the use of MRD testing in patients with AML.
Collapse
Affiliation(s)
- James S. Blachly
- Division of Hematology/Department of Medicine, The Ohio State University - The James Comprehensive Cancer Center, Columbus, OH,Department of Biomedical Informatics, The Ohio State University, Columbus, OH,J.S. Blachly
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA,Division of Hematology/Department of Medicine, University of Washington, Seattle, WA,Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA,Department of Epidemiology, 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, USA
| |
Collapse
|
77
|
Rodríguez-Arbolí E, Orvain C, Othus M, Walter RB. Significance of measurable residual disease in adults with secondary acute myeloid leukemia undergoing allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 2022; 57:1732-1734. [PMID: 36030308 PMCID: PMC10598788 DOI: 10.1038/s41409-022-01794-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Eduardo Rodríguez-Arbolí
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), University of Seville, Seville, Spain
| | - Corentin Orvain
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Megan Othus
- Public Health Science Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA.
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
78
|
Sahasrabudhe K, Huang Y, Rebechi M, Elder P, Mims A, Wall S. Survival, response rates, and post-transplant outcomes in patients with Acute Myeloid Leukemia aged 60-75 treated with high intensity chemotherapy vs. lower intensity targeted therapy. Front Oncol 2022; 12:1017194. [PMID: 36263213 PMCID: PMC9574198 DOI: 10.3389/fonc.2022.1017194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 12/01/2022] Open
Abstract
The United States Food and Drug Administration has approved several oral, targeted therapies for the treatment of Acute Myeloid Leukemia (AML) in recent years. These agents are approved in patients with relapsed/refractory disease or as frontline therapy in patients who are ineligible for intensive chemotherapy based on age, performance status, or comorbidities. They are also being increasingly utilized frontline in patients of all ages and fitness levels through clinical trials and off label prescribing, but comparative treatment outcomes associated with intensive versus targeted therapy have not been extensively studied. We conducted a single center, retrospective analysis to address the impact of treatment intensity on survival in patients with AML aged 60-75 at diagnosis. This study included 127 patients, 73 of whom received high intensity chemotherapy at any point during treatment (any HiC) and 54 of whom received only low intensity targeted therapy (LITT only). Overall survival (OS) from treatment initiation did not differ significantly between the any HiC and LITT only groups (hazard ratio (HR) for death, 0.67; 95% CI, 0.41 to 1.09; P=0.11). The only three variables that were independently associated with superior OS were lower European Leukemia Net (ELN) risk classification, TP53 unmutated status, and receipt of transplant. Our data suggest that baseline genomic features and receipt of transplant are more important than treatment intensity in predicting survival in this patient population. They also highlight the vital role of transplant in older patients with AML regardless of treatment intensity utilized for remission induction. Larger studies are needed to further address this question, including prospective randomized trials.
Collapse
Affiliation(s)
- Kieran Sahasrabudhe
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Ying Huang
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Melanie Rebechi
- Division of General Internal Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Patrick Elder
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Alice Mims
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Sarah Wall
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- *Correspondence: Sarah Wall,
| |
Collapse
|
79
|
Stone RM. Consolidation chemotherapy in AML: Are we playing with a full deck of cards? Best Pract Res Clin Haematol 2022; 35:101408. [DOI: 10.1016/j.beha.2022.101408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
80
|
Guolo F, Cerchione C, Vernarecci C, Isidori A. Editorial: Acute myeloid leukemia (AML): Is it time for MRD-driven treatment? Front Oncol 2022; 12:1020185. [PMID: 36158678 PMCID: PMC9502001 DOI: 10.3389/fonc.2022.1020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Fabio Guolo
- Clinic Of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
- Dipartimento di Oncologia ed Ematologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- *Correspondence: Fabio Guolo,
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Italy
| | - Chiara Vernarecci
- Clinic Of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Alessandro Isidori
- Haematology and Stem Cell Transplant Center, Azienda Ospedaliera Ospedali Riuniti Marche Nord (AORMN), Pesaro, Italy
| |
Collapse
|
81
|
Pessach I, Spyropoulos T, Lamprianidou E, Kotsianidis I. MRD Monitoring by Multiparametric Flow Cytometry in AML: Is It Time to Incorporate Immune Parameters? Cancers (Basel) 2022; 14:cancers14174294. [PMID: 36077826 PMCID: PMC9454571 DOI: 10.3390/cancers14174294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Measurable residual disease (MRD) is emerging as an important prognostic and predictive biomarker in acute myeloid leukemia (AML). However, its use is currently hampered by the disparity and lack of harmonization between the available MRD methodologies. In addition, the current assessment of MRD in AML focuses only on the quantification of the residual leukemic burden, without addressing the parallel alterations of the antineoplastic immune response that can critically affect the course and outcome of AML, often despite MRD persistence. Incorporating parameters of immune competence provides more consistency with the biological concept of MRD and may lead to higher accuracy. Multiparameter flow cytometry (MFC) is a highly efficacious and sensitive technology for the thorough and synchronous investigation of the kinetics of both antitumor immunity and the leukemic clone. MFC-based MRD provides the platform for the development of a composite leukemia- and immune-based biomarker which can outcompete the current MRD assessment. Abstract Acute myeloid leukemia (AML) is a heterogeneous group of clonal myeloid disorders characterized by intrinsic molecular variability. Pretreatment cytogenetic and mutational profiles only partially inform prognosis in AML, whereas relapse is driven by residual leukemic clones and mere morphological evaluation is insensitive for relapse prediction. Measurable residual disease (MRD), an independent post-diagnostic prognosticator, has recently been introduced by the European Leukemia Net as a new outcome definition. However, MRD techniques are not yet standardized, thus precluding its use as a surrogate endpoint for survival in clinical trials and MRD-guided strategies in real-life clinical practice. AML resistance and relapse involve a complex interplay between clonal and immune cells, which facilitates the evasion of the leukemic clone and which is not taken into account when merely quantifying the residual leukemia. Multiparameter flow cytometry (MFC) offers the possibility of capturing an overall picture of the above interactions at the single cell level and can simultaneously assess the competence of anticancer immune response and the levels of residual clonal cells. In this review, we focus on the current status of MFC-based MRD in diverse AML treatment settings and introduce a novel perspective of combined immune and leukemia cell profiling for MRD assessment in AML.
Collapse
Affiliation(s)
- Ilias Pessach
- Department of Hematology, Athens Medical Center, 11634 Athens, Greece
| | - Theodoros Spyropoulos
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 69100 Alexandroupolis, Greece
| | - Eleftheria Lamprianidou
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 69100 Alexandroupolis, Greece
| | - Ioannis Kotsianidis
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 69100 Alexandroupolis, Greece
- Correspondence: or ; Tel.: +30-25-5103-0320; Fax: +30-25-5107-6154
| |
Collapse
|
82
|
Lueck C, Panagiota V, Dammann E, Gabdoulline R, Berliner D, Veltmann C, Heuser M, Beutel G, Ganser A, Eder M. Increased Late Noncardiac Nonrelapse Mortality in Patients with Atrial Fibrillation Diagnosed During Their Hospital Stay for Allogeneic Stem Cell Transplantation. Transplant Cell Ther 2022; 28:609.e1-609.e8. [DOI: 10.1016/j.jtct.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/17/2022] [Accepted: 06/14/2022] [Indexed: 10/18/2022]
|
83
|
Bai L, Zhang YZ, Yan CH, Wang Y, Xu LP, Zhang XH, Zhang LP, Huang XJ, Cheng YF. Outcomes of allogeneic haematopoietic stem cell transplantation for paediatric patients with MLL-rearranged acute myeloid leukaemia. BMC Cancer 2022; 22:896. [PMID: 35974319 PMCID: PMC9382754 DOI: 10.1186/s12885-022-09978-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 08/01/2022] [Indexed: 11/10/2022] Open
Abstract
Background The presence of mixed-lineage leukaemia rearrangement (MLL-r) in paediatric patients with acute myeloid leukaemia (AML) is a poor prognostic predictor. Whether allogeneic haematopoietic stem cell transplantation (allo-HSCT) is beneficial in such cases remains unclear. Methods We evaluated the outcomes and prognostic factors of allo-HSCT in 44 paediatric patients with MLL-r AML in the first complete remission (CR1) between 2014 and 2019 at our institution. Results For all the 44 patients, the 3-year overall survival (OS), event-free survival (EFS), and cumulative incidence of relapse (CIR) were 74.5%, 64.1%, and 29.1%, respectively. Among them, 37 (84.1%) patients received haploidentical (haplo)-HSCT, and the 3-year OS, EFS, and CIR were 73.0%, 65.6%, and 26.4%, respectively. The 100-day cumulative incidence of grade II–IV acute graft-versus-host disease (aGVHD) post-transplantation was 27.3%, and that of grade III–IV aGVHD was 15.9%. The overall 3-year cumulative incidence of chronic graft-versus-host disease (cGVHD) post-transplantation was 40.8%, and that of extensive cGVHD was 16.7%. Minimal residual disease (MRD)-positive (MRD +) status pre-HSCT was significantly associated with lower survival and higher risk of relapse. The 3-year OS, EFS, and CIR differed significantly between patients with MRD + pre-HSCT (n = 15; 48.5%, 34.3% and 59%) and those with MRD-pre-HSCT (n = 29; 89.7%, 81.4% and 11.7%). Pre-HSCT MRD + status was an independent risk factor in multivariate analysis. Conclusions Allo-HSCT (especially haplo-HSCT) can be a viable strategy in these patients, and pre-HSCT MRD status significantly affected the outcomes.
Collapse
Affiliation(s)
- Lu Bai
- Department of Hematology, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Yong-Zhan Zhang
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Chen-Hua Yan
- Department of Hematology, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Yu Wang
- Department of Hematology, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Lan-Ping Xu
- Department of Hematology, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Xiao-Hui Zhang
- Department of Hematology, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Le-Ping Zhang
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Department of Hematology, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Yi-Fei Cheng
- Department of Hematology, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
84
|
Nayak RK, Chen YB. Maintenance therapy for AML after allogeneic HCT. Front Oncol 2022; 12:895771. [PMID: 36016625 PMCID: PMC9397403 DOI: 10.3389/fonc.2022.895771] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Allogeneic hematopoietic cell transplant (allo-HCT) for eligible patients with acute myeloid leukemia (AML) in first complete remission is a central treatment paradigm to achieve durable remission. However, disease relapse after allo-HCT remains a significant concern and generally portends a poor prognosis. There is significant interest regarding the role for maintenance therapy after allo-HCT for patients with high risk of relapse, regardless of the presence of measurable residual disease. While there are currently no therapies approved for maintenance therapy for AML after allo-HCT, there are a number of ongoing investigations examining the role of maintenance therapies that include targeted agents against FLT3-ITD or IDH mutations, hypomethylating agents, immunomodulatory therapies and cellular therapies. In this review, we examine the current landscape and future strategies for maintenance therapy for AML after allo-HCT.
Collapse
Affiliation(s)
- Rahul K. Nayak
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Yi-Bin Chen
- Hematopoietic Cell Transplant and Cell Therapy Program, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
85
|
El Chaer F, Ballen KK. Measurable residual disease for secondary acute myeloid leukemia prior to allogeneic hematopoietic cell transplantation: does it make a difference? Bone Marrow Transplant 2022; 57:1473-1474. [PMID: 35902643 DOI: 10.1038/s41409-022-01765-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Firas El Chaer
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Karen K Ballen
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
86
|
Bernardi M, Ferrara F, Carrabba MG, Mastaglio S, Lorentino F, Vago L, Ciceri F. MRD in Venetoclax-Based Treatment for AML: Does it Really Matter? Front Oncol 2022; 12:890871. [PMID: 35924144 PMCID: PMC9339596 DOI: 10.3389/fonc.2022.890871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
The prognosis of newly diagnosed patients with acute myeloid leukemia is still unfavorable in the majority of cases within the intermediate and mainly adverse genetic risk group but also in a considerable fraction of favorable-risk patients, mainly due to recurrence of disease after complete remission achievement or, less frequently, primary refractoriness. Besides genetic classification at diagnosis, post-treatment prognostic factors include measurable residual disease evaluation in patients in complete remission and in most cases measurable residual disease (MRD) positivity predicts hematologic relapse potentially allowing early therapeutic intervention. Currently, the most commonly used methods for detection of minimal residual disease are multiparameter flow cytometry and quantitative PCR, applicable to around 90% and 50% of patients, respectively. In addition, in > 90% of acute myeloid leukemia (AML) patients, molecular aberrations can be identified by next-generation sequencing, a technology that is widely used in clinical practice for the initial mutational screening at the time of diagnosis but more often, for MRD detection because its flexibility allows almost every mutated gene to be used as an MRD marker. Threshold levels of residual disease and correlation with outcome have been thoroughly studied and established in younger patients treated with intensive induction and consolidation chemotherapy as well as after allogeneic transplantation. Yet, experience on MRD monitoring and interpretation in patients treated with low-intensity regimens, including new agents, is still limited. The updated armamentarium of anti-leukemic agents includes the BCL-2 inhibitor venetoclax, which demonstrated good tolerability, high response rates, and prolonged overall survival when combined with hypomethylating agents or low dose cytarabine in patients considered elderly/”unfit” to tolerate intensive regimens. Although remissions with negative minimal residual disease clearly translated into improved outcomes after intensive treatments, data supporting the same evidence in patients receiving low-intensity venetoclax-based treatments are not still consolidated. We here review and discuss more recent data on the minimal residual disease interpretation and role in AML patients treated with venetoclax-based combinations.
Collapse
Affiliation(s)
- Massimo Bernardi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Department of Onco-Hematology, Milan, Italy
- *Correspondence: Massimo Bernardi,
| | | | - Matteo Giovanni Carrabba
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Department of Onco-Hematology, Milan, Italy
| | - Sara Mastaglio
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Department of Onco-Hematology, Milan, Italy
| | - Francesca Lorentino
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Department of Onco-Hematology, Milan, Italy
| | - Luca Vago
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Department of Onco-Hematology, Milan, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Fabio Ciceri
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Department of Onco-Hematology, Milan, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
87
|
Measurable residual disease (MRD) status before allogeneic hematopoietic cell transplantation impact on secondary acute myeloid leukemia outcome. A Study from the Acute Leukemia Working Party (ALWP) of the European society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant 2022; 57:1556-1563. [PMID: 35835997 DOI: 10.1038/s41409-022-01748-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 12/21/2022]
Abstract
Measurable residual disease (MRD) assessment before allogeneic hematopoietic cell transplantation (HCT) may help physicians to identify a subgroup of patients at high risk of relapse for de novo acute myeloid leukemia (AML) but its relevance among patients affected by secondary AML (sAML) is still unknown. We assessed the impact of MRD among 318 adult patients with sAML who received an allogeneic HCT in first complete remission. At the time of HCT, a total of 208 (65%) patients achieved MRD negativity, while 110 (35%) had positive MRD. 2-year overall survival (OS) was 58.8 % (95% CI 52.2-64.9) with leukemia-free survival (LFS) of 50.0 % (95% CI 43.7-56.1), relapse incidence of 34.2% (95% CI 28.4-40.1) and non-relapse mortality (NRM) of 23.3 % (95% CI 19-27.7) for the entire cohort. In multivariate analysis, HCT recipients with KPS ≥ 90 experienced less disease recurrence (HR 0.61, 95% CI 0.4-0.94) with better LFS (HR 0.63, 95% CI 0.44-0.89) and OS (HR 0.58, 95% CI 0.39-0.86). There were no differences in major clinical endpoints between patients with MRD-positive and MRD-negative status at the time of HCT. Pre-transplantation assessment of MRD was not informative on post-HCT outcomes in this retrospective registry-based analysis among patients affected by sAML.
Collapse
|
88
|
Winters AC, Bosma G, Abbott D, Minhajuddin M, Jordan C, Pollyea DA, Gutman JA. Outcomes Are Similar After Allogeneic Hematopoietic Stem Cell Transplant for Newly Diagnosed Acute Myeloid Leukemia Patients who Received Venetoclax + Azacitidine Versus Intensive Chemotherapy. Transplant Cell Ther 2022; 28:694.e1-694.e9. [DOI: 10.1016/j.jtct.2022.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/05/2022] [Accepted: 07/20/2022] [Indexed: 10/16/2022]
|
89
|
Short NJ, Kantarjian H. Choosing between intensive and less intensive front-line treatment approaches for older patients with newly diagnosed acute myeloid leukaemia. Lancet Haematol 2022; 9:e535-e545. [PMID: 35772432 DOI: 10.1016/s2352-3026(22)00167-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
The outcomes of older patients with acute myeloid leukaemia are inferior to their younger counterparts, because, in part, of a more aggressive disease biology and poorer tolerance of cytotoxic chemotherapy. Although intensive chemotherapy was historically considered the only effective treatment for these patients, many older patients are not suitable for intensive chemotherapy owing to comorbidities or general frailty. Determination of patient fitness for intensive chemotherapy is imperfect, and even older patients who appear to be suitable (also known as fit) for intensive chemotherapy can have high rates of morbidity and early and late mortality with this approach. Fortunately, the outcomes of older or unfit patients with acute myeloid leukaemia have substantially improved with the use of a hypomethylating agent plus venetoclax in the front-line setting. Although the formal approval of this combination is limited to patients aged 75 years or older, or those with a clinically significant comorbidity, the high response rates and survival improvement in these patients have led many practitioners to consider this low-intensity regimen in older patients without significant comorbidities and even in younger patients with high-risk disease features for whom the expected outcomes with intensive chemotherapy are poor. Modifications to the hypomethylating agent plus venetoclax backbone might further improve the outlook for these patients, particularly in some acute myeloid leukaemia subsets with a targetable mutation. In this Viewpoint, we review the retrospective and prospective data supporting both intensive chemotherapy and low-intensity venetoclax-based approaches in older patients with acute myeloid leukaemia. We also discuss our own approach to the management of older or unfit patients with acute myeloid leukaemia, including how cytomolecular features have a role in establishing the optimal front-line therapy.
Collapse
Affiliation(s)
- Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
90
|
Molecular Measurable Residual Disease Assessment before Hematopoietic Stem Cell Transplantation in Pediatric Acute Myeloid Leukemia Patients: A Retrospective Study by the I-BFM Study Group. Biomedicines 2022; 10:biomedicines10071530. [PMID: 35884834 PMCID: PMC9313005 DOI: 10.3390/biomedicines10071530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/14/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a curative post-remission treatment in patients with acute myeloid leukemia (AML), but relapse after transplant is still a challenging event. In recent year, several studies have investigated the molecular minimal residual disease (qPCR-MRD) as a predictor of relapse, but the lack of standardized protocols, cut-offs, and timepoints, especially in the pediatric setting, has prevented its use in several settings, including before HSCT. Here, we propose the first collaborative retrospective I-BFM-AML study assessing qPCR-MRD values in pretransplant bone marrow samples of 112 patients with a diagnosis of AML harboring t(8;21)(q22; q22)RUNX1::RUNX1T1, or inv(16)(p13q22)CBFB::MYH11, or t(9;11)(p21;q23)KMT2A::MLLT3, or FLT3-ITD genetic markers. We calculated an ROC cut-off of 2.1 × 10−4 that revealed significantly increased OS (83.7% versus 57.1%) and EFS (80.2% versus 52.9%) for those patients with lower qPCR-MRD values. Then, we partitioned patients into three qPCR-MRD groups by combining two different thresholds, 2.1 × 10−4 and one lower cut-off of 1 × 10−2, and stratified patients into low-, intermediate-, and high-risk groups. We found that the 5-year OS (83.7%, 68.6%, and 39.2%, respectively) and relapse-free survival (89.2%, 73.9%, and 67.9%, respectively) were significantly different independent of the genetic lesion, conditioning regimen, donor, and stem cell source. These data support the PCR-based approach playing a clinical relevance in AML transplant management.
Collapse
|
91
|
Wang L, Chen R, Li L, Zhu L, Huang X, Ye X. Prognostic implication of early minimal residual disease evaluation in patients with chronic myelomonocytic leukemia. Am J Cancer Res 2022; 12:2216-2225. [PMID: 35693086 PMCID: PMC9185623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 04/15/2022] [Indexed: 06/15/2023] Open
Abstract
To investigate the prognostic implication of minimal residual disease (MRD) evaluation in chronic myelomonocytic leukemia (CMML), we conducted a restropective study included a total of 174 CMML patients in our hospital from January 2010 to March 2021. In which 50/174 (29%) bone marrow samples were conducted by multiparameter flow cytometry (FCM) assessed MRD analysis after the first three cycles of treatment and were included in this study. MRD was detected by six- to eight-colour FCM. The achievement of early MRD negativity had better clinical outcomes in patients with CMML, which fared better prognosis in terms of not only PFS (P=0.006) but also OS (P=0.02) after the first cycle, and PFS (P=0.023 and P=0.041) after the second and third cycles, whereas no significantly influence in OS. In addition, MRD negative after initial treatment remained its independent prognostic value associated with PFS (adjusted hazard ratio [HR] 0.161, 95 CI 0.035-0.738; P=0.019) and OS (adjusted HR 0.136; 95 CI 0.017-1.077; P=0.059), indicating that patients with MRD-negative after the initial treatment alone could obtain the greatest clinical benefit. According to MRD level, the patients were divided into 4 different groups: very low risk (fewer than 10-4 cells) in 15 cases, low risk (10-4 to 10-3 cells) in 6; and 6 were at intermediate risk (fewer than 10-3 to 10-2 cells). The rest of 23 patients were were assigned to the high-risk grades (more than 10-2 residual cells), we find this risk stratification model is significantly associated with better PFS (P=0.002) but marginal significantly associated with OS (P=0.068). Notably, patients with DNMT3A mutation fared a shorter PFS in the MRD positive subgroup (P=0.068). MRD is highly predictive of prognosis, and its combination with molecular profile may help identify patients at increased risk for progression to further improve the management of patients with CMML. Large-scaled investigations are warranted to validate our conclusions and its potential in clinical practice.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
- Program in Clinical Medicine, School of Medicine of Zhejiang UniversityHangzhou, Zhejiang, China
| | - Rongrong Chen
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
- Program in Clinical Medicine, School of Medicine of Zhejiang UniversityHangzhou, Zhejiang, China
| | - Li Li
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Lixia Zhu
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Xianbo Huang
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Xiujin Ye
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| |
Collapse
|
92
|
Sahasrabudhe KD, Mims AS. Venetoclax and acute myeloid leukaemia: an expanding new frontier. THE LANCET HAEMATOLOGY 2022; 9:e387-e389. [DOI: 10.1016/s2352-3026(22)00136-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/26/2022]
|
93
|
Paras G, Morsink LM, Othus M, Milano F, Sandmaier BM, Zarling LC, Palmieri R, Schoch G, Davis C, Bleakley M, Flowers MED, Deeg HJ, Appelbaum FR, Storb R, Walter RB. Conditioning intensity and peritransplant flow cytometric MRD dynamics in adult AML. Blood 2022; 139:1694-1706. [PMID: 34995355 PMCID: PMC8931514 DOI: 10.1182/blood.2021014804] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/28/2021] [Indexed: 11/20/2022] Open
Abstract
In acute myeloid leukemia (AML), measurable residual disease (MRD) before or after allogeneic hematopoietic cell transplantation (HCT) is an established independent indicator of poor outcome. To address how peri-HCT MRD dynamics could refine risk assessment across different conditioning intensities, we analyzed 810 adults transplanted in first or second remission after myeloablative conditioning (MAC; n = 515) or non-MAC (n = 295) who underwent multiparameter flow cytometry-based MRD testing before as well as 20 to 40 days after allografting. Patients without pre- and post-HCT MRD (MRDneg/MRDneg) had the lowest risks of relapse and highest relapse-free survival (RFS) and overall survival (OS). Relative to those patients, outcomes for MRDpos/MRDpos and MRDneg/MRDpos patients were poor regardless of conditioning intensity. Outcomes for MRDpos/MRDneg patients were intermediate. Among 161 patients with MRD before HCT, MRD was cleared more commonly with a MAC (85 of 104; 81.7%) than non-MAC (33 of 57; 57.9%) regimen (P = .002). Although non-MAC regimens were less likely to clear MRD, if they did, the impact on outcome was greater. Thus, there was a significant interaction between conditioning intensity and "MRD conversion" for relapse (P = .020), RFS (P = .002), and OS (P = .001). Similar findings were obtained in the subset of 590 patients receiving HLA-matched allografts. C-statistic values were higher (indicating higher predictive accuracy) for peri-HCT MRD dynamics compared with the isolated use of pre-HCT MRD status or post-HCT MRD status for prediction of relapse, RFS, and OS. Across conditioning intensities, peri-HCT MRD dynamics improve risk assessment over isolated pre- or post-HCT MRD assessments in patients with AML.
Collapse
Affiliation(s)
- Gabrielle Paras
- Department of Medicine, Residency Program, University of Washington, Seattle, WA
| | - Linde M Morsink
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Hematology, University Medical Center Groningen, Groningen, The Netherlands
| | - Megan Othus
- Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Filippo Milano
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Brenda M Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Lucas C Zarling
- Department of Medicine, Residency Program, University of Washington, Seattle, WA
| | - Raffaele Palmieri
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Hematology, University Tor Vergata, Rome, Italy; and
| | - Gary Schoch
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Chris Davis
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Pediatrics
| | - Mary E D Flowers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - H Joachim Deeg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Frederick R Appelbaum
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Rainer Storb
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Hematology, Department of Medicine
- Department of Laboratory Medicine & Pathology, and
- Department of Epidemiology, University of Washington, Seattle, WA
| |
Collapse
|
94
|
Cluzeau T, Lemoli RM, McCloskey J, Cooper T. Measurable Residual Disease in High-Risk Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:cancers14051278. [PMID: 35267586 PMCID: PMC8909238 DOI: 10.3390/cancers14051278] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Assessment of measurable residual disease (MRD) identifies small numbers of acute myeloid leukemia (AML) cells that may remain after initiating treatment. The achievement of MRD negativity (no detectable AML cells remaining) typically predicts better outcomes for patients with AML. Some patients with AML have disease characteristics that put them at a higher risk of treatment failure or relapse; while outcomes for patients with high-risk AML are historically poor with traditional chemotherapy regimens, newer chemotherapy formulations (i.e., CPX-351) and targeted therapies may be more effective in achieving MRD negativity in these patients. Currently, there is no agreement on the best method for determining whether a patient has achieved MRD negativity, and the use of several different methods makes it difficult to compare outcomes across studies. Despite these challenges, regular monitoring of patients for the achievement of MRD negativity will become increasingly important in the routine management of patients with high-risk AML. Abstract Mounting evidence suggests measurable residual disease (MRD) assessments are prognostic in acute myeloid leukemia (AML). High-risk AML encompasses a subset of AML with poor response to therapy and prognosis, with features such as therapy-related AML, an antecedent hematologic disorder, extramedullary disease (in adults), and selected mutations and cytogenetic abnormalities. Historically, few patients with high-risk AML achieved deep and durable remission with conventional chemotherapy; however, newer agents might be more effective in achieving MRD-negative remission. CPX-351 (dual-drug liposomal encapsulation of daunorubicin/cytarabine at a synergistic ratio) demonstrated MRD-negativity rates of 36–64% across retrospective studies in adults with newly diagnosed high-risk AML and 84% in pediatric patients with first-relapse AML. Venetoclax (BCL2 inhibitor) demonstrated MRD-negativity rates of 33–53% in combination with hypomethylating agents for high-risk subgroups in studies of older adults with newly diagnosed AML who were ineligible for intensive therapy and 65% in combination with chemotherapy in pediatric patients with relapsed/refractory AML. However, there is no consensus on optimal MRD methodology in AML, and the use of different techniques, sample sources, sensitivity thresholds, and the timing of assessments limit comparisons across studies. Robust MRD analyses are needed in future clinical studies, and MRD monitoring should become a routine aspect of AML management.
Collapse
Affiliation(s)
- Thomas Cluzeau
- Service d’hématologie, Université Cote d’Azur, CHU de Nice, 06200 Nice, France
- Correspondence: ; Tel.: +33-492035841; Fax: +33-492035895
| | - Roberto M. Lemoli
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
- Clinic of Hematology, Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| | - James McCloskey
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA;
| | - Todd Cooper
- Division of Hematology/Oncology, Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA 98105, USA;
| |
Collapse
|
95
|
Leotta S, Condorelli A, Sciortino R, Milone GA, Bellofiore C, Garibaldi B, Schininà G, Spadaro A, Cupri A, Milone G. Prevention and Treatment of Acute Myeloid Leukemia Relapse after Hematopoietic Stem Cell Transplantation: The State of the Art and Future Perspectives. J Clin Med 2022; 11:253. [PMID: 35011994 PMCID: PMC8745746 DOI: 10.3390/jcm11010253] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) for high-risk acute myeloid leukemia (AML) represents the only curative option. Progress has been made in the last two decades in the pre-transplant induction therapies, supportive care, selection of donors and conditioning regimens that allowed to extend the HSCT to a larger number of patients, including those aged over 65 years and/or lacking an HLA-identical donor. Furthermore, improvements in the prophylaxis of the graft-versus-host disease and of infection have dramatically reduced transplant-related mortality. The relapse of AML remains the major reason for transplant failure affecting almost 40-50% of the patients. From 10 to 15 years ago to date, treatment options for AML relapsing after HSCT were limited to conventional cytotoxic chemotherapy and donor leukocyte infusions (DLI). Nowadays, novel agents and targeted therapies have enriched the therapeutic landscape. Moreover, very recently, the therapeutic landscape has been enriched by manipulated cellular products (CAR-T, CAR-CIK, CAR-NK). In light of these new perspectives, careful monitoring of minimal-residual disease (MRD) and prompt application of pre-emptive strategies in the post-transplant setting have become imperative. Herein, we review the current state of the art on monitoring, prevention and treatment of relapse of AML after HSCT with particular attention on novel agents and future directions.
Collapse
Affiliation(s)
| | - Annalisa Condorelli
- Division of Hematology, AOU “Policlinico G. Rodolico-San Marco”, Via Santa Sofia 78, 95124 Catania, Italy; (S.L.); (R.S.); (G.A.M.); (C.B.); (B.G.); (G.S.); (A.S.); (A.C.); (G.M.)
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Cohen J, Maziarz RT. Post-allogeneic stem cell transplant FLT3- targeted maintenance therapy: updates and considerations for clinical practice. ARCHIVES OF STEM CELL AND THERAPY 2022; 3:23-27. [PMID: 36594901 PMCID: PMC9802584 DOI: 10.46439/stemcell.3.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Acute myeloid leukemia (AML) is characterized by multiple molecular and cytogenetic abnormalities, with increasing data to support clinical and prognostic implications to guide clinical decision making. One of the most well described mutations involves fms-like tyrosine kinase 3 (FLT3) that results in a constitutively active tyrosine kinase and is generally associated with poor prognosis involving shorter overall survival and higher rates of relapse. Advancements in targeted therapies have greatly influenced available treatment options in a landscape that has remained largely unchanged for the past five decades. Tyrosine kinase inhibitors (TKI), specifically FLT3-targeted therapies, are now integral treatment options for patients with this targetable mutation. As allogeneic hematopoietic cell transplant (alloHCT) remains the primary curative therapy for most adult AML patients, the goal is for eligible patients to proceed to transplant. However, post-alloHCT relapse remains exceedingly high even in patients achieving deep responses to therapy. Limited evaluation of FLT3-targeted TKIs as post-alloHCT maintenance therapy in FLT3-positive patients suggest improved outcomes and tolerable safety profiles, with ongoing studies further investigating second-generation agents. Thus, this commentary aims to review the role of post-alloHCT FLT3-targeted maintenance therapy and considerations for clinical practice.
Collapse
Affiliation(s)
- Jonathan Cohen
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA,Department of Pharmacy, Oregon Health & Science University, Portland, OR USA,Author for correspondence:
| | - Richard T. Maziarz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA,Center for Hematologic Malignancies, Oregon Health & Science University, Portland, OR USA
| |
Collapse
|
97
|
Measurable Residual Disease Assessment as a Surrogate Marker in New Drug Development in Acute Myeloid Leukemia. Cancer J 2022; 28:73-77. [PMID: 35072377 PMCID: PMC8849520 DOI: 10.1097/ppo.0000000000000572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
ABSTRACT Response criteria for patients treated for acute myeloid leukemia (AML) based on cytomorphology are inadequate. Many patients achieving a complete remission by such criteria will later relapse. Patients with AML in such remissions who test negative using higher sensitivity measures of residual disease burden (measurable residual disease [MRD]) have on average lower relapse rates and better survival than those testing positive. This association has raised the possibility that these technological advances in measurement of tumor burden could be used to optimize the drug development and regulatory approval processes in AML. The heterogeneous genetic etiology, diverse immunophenotypic profiles, related precursor states and polyclonal architecture however combine to make the development of standardized and validated MRD assessments for AML challenging. Current and future methods to measure residual disease in AML, performance characteristics of testing currently in use, and potential uses for optimized AML MRD tests including as a surrogate endpoint are discussed.
Collapse
|
98
|
The Evolving Role of Allogeneic Stem Cell Transplant in the Era of Molecularly Targeted Agents. Cancer J 2022; 28:78-84. [DOI: 10.1097/ppo.0000000000000575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
99
|
Technical Aspects of Flow Cytometry-based Measurable Residual Disease Quantification in Acute Myeloid Leukemia: Experience of the European LeukemiaNet MRD Working Party. Hemasphere 2022; 6:e676. [PMID: 34964040 PMCID: PMC8701786 DOI: 10.1097/hs9.0000000000000676] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Measurable residual disease (MRD) quantified by multiparameter flow cytometry (MFC) is a strong and independent prognostic factor in acute myeloid leukemia (AML). However, several technical factors may affect the final read-out of the assay. Experts from the MRD Working Party of the European LeukemiaNet evaluated which aspects are crucial for accurate MFC-MRD measurement. Here, we report on the agreement, obtained via a combination of a cross-sectional questionnaire, live discussions, and a Delphi poll. The recommendations consist of several key issues from bone marrow sampling to final laboratory reporting to ensure quality and reproducibility of results. Furthermore, the experiences were tested by comparing two 8-color MRD panels in multiple laboratories. The results presented here underscore the feasibility and the utility of a harmonized theoretical and practical MFC-MRD assessment and are a next step toward further harmonization.
Collapse
|
100
|
Zhao C, Sun YQ, Xu LP, Zhang XH, Liu KY, Huang XJ, Wang Y. [Impact of blood count recovery before haploidentical stem cell transplantation on outcomes in acute myeloid leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:1030-1034. [PMID: 35045676 PMCID: PMC8770879 DOI: 10.3760/cma.j.issn.0253-2727.2021.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Indexed: 11/05/2022]
Affiliation(s)
- C Zhao
- Peking University People' s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Y Q Sun
- Peking University People' s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - L P Xu
- Peking University People' s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - X H Zhang
- Peking University People' s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - K Y Liu
- Peking University People' s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - X J Huang
- Peking University People' s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China Hematology Collaborative Innovation Center, Peking University, Beijing 100044, China
| | - Y Wang
- Peking University People' s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China Hematology Collaborative Innovation Center, Peking University, Beijing 100044, China
| |
Collapse
|