1
|
Liu J, Guo D, Lian H, Ding P, Liu X, Zhao Y, Li H, Fan S. Combination of pre-transplant flow cytometry, WT1 expression, and NGS for MRD monitoring is potent in predicting the prognosis of AML receiving allogeneic transplantation. Ann Hematol 2025:10.1007/s00277-025-06384-0. [PMID: 40293465 DOI: 10.1007/s00277-025-06384-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
Minimal residual disease (MRD) monitoring has been demonstrated to important in predicting prognosis in acute myeloid leukemia (AML) receiving allogeneic hematopoietic stem cell transplantation (allo-HSCT), but the ideal time point and method remain unclear. Our study compared the prognostic value of multiparameter flow cytometry (MFC)-based and WT1 expression-based MRD a month before allo-HSCT [HSCT(-1 m)] and after allo-HSCT [HSCT(+ 1 m)], as well as next generation sequencing (NGS)-based MRD at HSCT(-1 m), HSCT(+ 1 m), 3 and 6 months after allo-HSCT [HSCT(+ 3 m) and HSCT(+ 6 m)] among 47 AML patients undergoing allo-HSCT. The MRD status by all the methods at HSCT(-1 m) was proved as a superior indicator with prognostic significance for disease progression, compared to that at HSCT(+ 1 m). For the NGS-based MRD, HSCT(+ 6 m) seemed to be the optimal detection time point, as supported by the optimal prognostic discrimination capability and the relatively high sensitivity for disease progression prediction. Moreover, our data showed that each individual method had some limitations in predicting prognosis; however, pre-transplant MRD monitoring by the combination of MFC, WT1 and NGS could greatly increase the sensitivity (100%) of identifying disease progression and greatly improve prognostic stratification. Our study may provide insights into the optimal time point and methodology for MRD monitoring in AML following allo-HSCT.
Collapse
Affiliation(s)
- Jie Liu
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Dan Guo
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Hanxi Lian
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Peiwen Ding
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Xin Liu
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Yanqiu Zhao
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Huibo Li
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Shengjin Fan
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China.
- NHC Key Laboratory of Cell Transplantation, the First Affiliated Hospital, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
2
|
Jum’ah HA, Otteson GE, Timm MM, Weybright MJ, Shi M, Horna P, Jevremovic D, Reichard KK, Olteanu H. Measurable Residual Disease Analysis by Flow Cytometry: Assay Validation and Characterization of 385 Consecutive Cases of Acute Myeloid Leukemia. Cancers (Basel) 2025; 17:1155. [PMID: 40227672 PMCID: PMC11987847 DOI: 10.3390/cancers17071155] [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: 01/31/2025] [Revised: 03/07/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Background/Objectives: Acute myeloid leukemia (AML) is a biologically heterogeneous malignancy with a variable prognosis. Despite many patients achieving complete remission, relapse remains common, underscoring the need for effective prognostic markers. Measurable residual disease (MRD) has emerged as a critical prognostic indicator, associated with higher relapse risk and shorter survival. This study reports on our initial experience of MRD detection by flow cytometry in 385 bone marrow samples from 126 AML patients. Methods: The flow cytometry MRD assay, validated according to stringent consensus recommendations, consists of a 3-tube, 10-color panel incorporating a broad spectrum of lineage differentiation markers. Analytical specificity, sensitivity, precision, and reproducibility were evaluated, demonstrating the assay's robustness. Results: The results reveal distinct immunophenotypic aberrancies in all AML cases, with consistent identification of aberrant immunophenotypes in follow-up specimens. AML MRD was detected in 32 out of 126 patients (25%) and in 77 out of 385 analyses (20%), with a median aberrant blast percentage of 1.87% (range, 0.01-12). A change in immunophenotype was documented in 21% of the MRD-positive cases. MRD positivity detected in the first sample studied was associated with reduced overall survival (HR: 5.153; p < 0.0001). Conclusions: Our findings support the integration of flow cytometric MRD analysis into routine clinical practice to enhance risk stratification and treatment planning for AML patients, as currently recommended by professional guidelines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Horatiu Olteanu
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN 55905, USA; (H.A.J.)
| |
Collapse
|
3
|
Cai Q, Lan H, Yi D, Xian B, Zidan L, Li J, Liao Z. Flow cytometry in acute myeloid leukemia and detection of minimal residual disease. Clin Chim Acta 2025; 564:119945. [PMID: 39209245 DOI: 10.1016/j.cca.2024.119945] [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: 06/28/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Acute myeloid leukemia (AML) is a common type of acute leukemia (AL), belonging to malignant tumors of the hematopoietic system with the characteristics of rapid disease development, control with extreme difficulties, easy recurrence, poor prognosis, and incidence rate increasing with age. The traditionally diagnostic standard of French American British (FAB), being based on the morphological examination with high human subjectivity, can no longer meet the demand of clinical diagnosis and treatment of AML. Requirements of objective accuracy and low-dose sample, have become the indispensable method for AML diagnosis and monitoring prognosis. Flow cytometry is a modern technology that can quickly and accurately detect the series, antigen distribution, differentiation stage of AML cells, minimal residual lesions after AML therapy, so as to provide the great significance in guiding clinical diagnosis, hierarchical treatment, and prognosis judgement. This article will systematically elaborate on the application of flow cytometry in the diagnosis and classification of AML, and the detection of minimal residual lesions, thereby providing reference significance for dynamic monitoring and prognostic observation of AML with different immune subtypes of FAB.
Collapse
Affiliation(s)
- Qihui Cai
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Haiqiang Lan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Deng Yi
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Bojun Xian
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Luo Zidan
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Jianqiao Li
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Zhaohong Liao
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
4
|
Zhao Z, Lan J. Detection methods and prognosis implications of measurable residual disease in acute myeloid leukemia. Ann Hematol 2024; 103:4869-4881. [PMID: 39283479 DOI: 10.1007/s00277-024-06008-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/11/2024] [Indexed: 01/16/2025]
Abstract
Measurable residual disease (MRD) in acute myeloid leukemia (AML) refers to the quantity of residual leukemic cells in a patient after treatment.According to the latest agreements, MRD in AML offering essential prognostic insights. However, there is ongoing debate regarding MRD-based monitoring and treatment strategies. There are multiple platforms for detecting MRD, each varying in sensitivity and suitability for different patients. MRD not only predicts treatment outcomes but also serves as an indicator of treatment effectiveness and a prognostic biomarker. In AML, most retrospective studies indicate that patients who are MRD-positive or show increasing MRD levels at specific time points during remission have significantly higher risks of relapse and mortality compared to MRD-negative patients. Although achieving MRD-negative status can improve patient prognosis, the possibility of relapse remains. Despite the correlation between MRD and clinical outcomes, MRD assessment methods are not yet standardized, leading to discrepancies in results across different techniques. To provide reliable MRD results, it is essential to optimize and standardize MRD detection methods. Methods for assessing MRD include multiparameter flow cytometry (MFC) and molecular assays, chosen based on disease characteristics. This review focuses on currently available MRD detection methods and discusses how the prognostic value of MRD test results informs personalized treatment strategies for AML patients.
Collapse
Affiliation(s)
- Zihan Zhao
- The Second Clinical Medical College, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jianping Lan
- Cancer Center, Department of Hematology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Zhejiang, Hangzhou, 310014, China.
| |
Collapse
|
5
|
Shen Q, Gong X, Feng Y, Hu Y, Wang T, Yan W, Zhang W, Qi S, Gale RP, Chen J. Measurable residual disease (MRD)-testing in haematological cancers: A giant leap forward or sideways? Blood Rev 2024; 68:101226. [PMID: 39164126 DOI: 10.1016/j.blre.2024.101226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024]
Abstract
Measurable residual disease (MRD)-testing is used in many haematological cancers to estimate relapse risk and to direct therapy. Sometimes MRD-test results are used for regulatory approval. However, some people including regulators wrongfully believe results of MRD-testing are highly accurate and of proven efficacy in directing therapy. We review MRD-testing technologies and evaluate the accuracy of MRD-testing for predicting relapse and the strength of evidence supporting efficacy of MRD-guided therapy. We show that at the individual level MRD-test results are often an inaccurate relapse predictor. Also, no convincing data indicate that increasing therapy-intensity based on a positive MRD-test reduces relapse risk or improves survival. We caution against adjusting therapy-intensity based solely on results of MRD-testing.
Collapse
Affiliation(s)
- Qiujin Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Xiaowen Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Yahui Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Tiantian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Wen Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Wei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Saibing Qi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK.
| | - Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| |
Collapse
|
6
|
Duployez N, Preudhomme C. Monitoring molecular changes in the management of myelodysplastic syndromes. Br J Haematol 2024; 205:772-779. [PMID: 38934371 DOI: 10.1111/bjh.19614] [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: 04/18/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
The ongoing or anticipated therapeutic advances as well as previous experience in other malignancies, including acute myeloid leukaemia, have made molecular monitoring a potential interesting tool for predicting outcomes and demonstrating treatment efficacy in patients with myelodysplastic syndromes (MDS). The important genetic heterogeneity in MDS has made challenging the establishment of recommendations. In this context, high-throughput/next-generation sequencing (NGS) has emerged as an attractive tool, especially in patients with high-risk diseases. However, its implementation in clinical practice still suffers from a lack of standardization in terms of sensitivity, bioinformatics and result interpretation. Data from literature, mostly gleaned from retrospective cohorts, show NGS monitoring when used appropriately could help clinicians to guide therapy, detect early relapse and predict disease evolution. Translating these observations into personalized patient management requires a prospective evaluation in clinical research and remains a major challenge for the next years.
Collapse
Affiliation(s)
- Nicolas Duployez
- Laboratory of Haematology, CHU Lille, Lille, France
- U1277 CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), University of Lille, INSERM, Lille, France
| | - Claude Preudhomme
- Laboratory of Haematology, CHU Lille, Lille, France
- U1277 CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), University of Lille, INSERM, Lille, France
| |
Collapse
|
7
|
Sun Y, Zhu G, Zhong H. Minimal residual disease monitoring in acute myeloid leukemia: Focus on MFC-MRD and treatment guidance for elderly patients. Eur J Haematol 2024; 112:870-878. [PMID: 38342613 DOI: 10.1111/ejh.14187] [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: 11/24/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/13/2024]
Abstract
Acute myeloid leukemia (AML) is distinguished by clonal growth of myeloid precursor cells, which impairs normal hematopoiesis. Minimal residual disease (MRD) refers to the residual leukemia cells that persist after chemotherapy. Patients who test positive for MRD have a higher likelihood of experiencing a recurrence, regardless of the specific chemotherapy approach used. Multi-parameter flow cytometry (MFC), polymerase chain reaction (PCR), and next-generation sequencing (NGS) are commonly employed techniques for identifying MRD. In the context of AML, patients are frequently monitored for measurable residual disease via multi-parameter flow cytometry (MFC-MRD). In order to explore recent advancements in AML and MRD diagnosis, an extensive search of the PubMed database was conducted, focusing on relevant research in the past 20 years. This review aims to examine various MRD monitoring methods, the optimal time points for assessment, as well as different specimen types used. Additionally, it underscores the significance of MFC-MRD assessment in guiding the treatment of elderly AML.
Collapse
Affiliation(s)
- Yue Sun
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Gelan Zhu
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Hua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
8
|
Modak RV, de Oliveira Rebola KG, McClatchy J, Mohammadhosseini M, Damnernsawad A, Kurtz SE, Eide CA, Wu G, Laderas T, Nechiporuk T, Gritsenko MA, Hansen JR, Hutchinson C, Gosline SJ, Piehowski P, Bottomly D, Short N, Rodland K, McWeeney SK, Tyner JW, Agarwal A. Targeting CCL2/CCR2 Signaling Overcomes MEK Inhibitor Resistance in Acute Myeloid Leukemia. Clin Cancer Res 2024; 30:2245-2259. [PMID: 38451486 PMCID: PMC11094423 DOI: 10.1158/1078-0432.ccr-23-2654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/29/2023] [Accepted: 03/05/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Emerging evidence underscores the critical role of extrinsic factors within the microenvironment in protecting leukemia cells from therapeutic interventions, driving disease progression, and promoting drug resistance in acute myeloid leukemia (AML). This finding emphasizes the need for the identification of targeted therapies that inhibit intrinsic and extrinsic signaling to overcome drug resistance in AML. EXPERIMENTAL DESIGN We performed a comprehensive analysis utilizing a cohort of ∼300 AML patient samples. This analysis encompassed the evaluation of secreted cytokines/growth factors, gene expression, and ex vivo drug sensitivity to small molecules. Our investigation pinpointed a notable association between elevated levels of CCL2 and diminished sensitivity to the MEK inhibitors (MEKi). We validated this association through loss-of-function and pharmacologic inhibition studies. Further, we deployed global phosphoproteomics and CRISPR/Cas9 screening to identify the mechanism of CCR2-mediated MEKi resistance in AML. RESULTS Our multifaceted analysis unveiled that CCL2 activates multiple prosurvival pathways, including MAPK and cell-cycle regulation in MEKi-resistant cells. Employing combination strategies to simultaneously target these pathways heightened growth inhibition in AML cells. Both genetic and pharmacologic inhibition of CCR2 sensitized AML cells to trametinib, suppressing proliferation while enhancing apoptosis. These findings underscore a new role for CCL2 in MEKi resistance, offering combination therapies as an avenue to circumvent this resistance. CONCLUSIONS Our study demonstrates a compelling rationale for translating CCL2/CCR2 axis inhibitors in combination with MEK pathway-targeting therapies, as a potent strategy for combating drug resistance in AML. This approach has the potential to enhance the efficacy of treatments to improve AML patient outcomes.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Receptors, CCR2/metabolism
- Receptors, CCR2/antagonists & inhibitors
- Receptors, CCR2/genetics
- Drug Resistance, Neoplasm/genetics
- Chemokine CCL2/metabolism
- Chemokine CCL2/genetics
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Signal Transduction/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Animals
- Pyridones/pharmacology
- Pyridones/therapeutic use
- Mice
Collapse
Affiliation(s)
- Rucha V. Modak
- Division of Oncological Sciences, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Katia G. de Oliveira Rebola
- Division of Oncological Sciences, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - John McClatchy
- Division of Oncological Sciences, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Mona Mohammadhosseini
- Division of Oncological Sciences, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Alisa Damnernsawad
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Stephen E. Kurtz
- Division of Oncological Sciences, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Christopher A. Eide
- Division of Oncological Sciences, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Guanming Wu
- Division of Bioinformatics & Computational Biology, Oregon Health & Science University, Portland, Oregon
| | - Ted Laderas
- Division of Bioinformatics & Computational Biology, Oregon Health & Science University, Portland, Oregon
| | - Tamilla Nechiporuk
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | | | | | | | - Sara J.C. Gosline
- Pacific Northwest National Laboratory, Richland, Washington
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Paul Piehowski
- Pacific Northwest National Laboratory, Richland, Washington
| | - Daniel Bottomly
- Division of Bioinformatics & Computational Biology, Oregon Health & Science University, Portland, Oregon
| | - Nicholas Short
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas
| | - Karin Rodland
- Pacific Northwest National Laboratory, Richland, Washington
| | - Shannon K. McWeeney
- Division of Bioinformatics & Computational Biology, Oregon Health & Science University, Portland, Oregon
| | - Jeffrey W. Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Anupriya Agarwal
- Division of Oncological Sciences, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
9
|
Yanada M, Yano S, Kuwatsuka Y, Kawamura K, Fukuda T, Ichinohe T, Hashii Y, Goto H, Kato K, Ishimaru F, Sato A, Onizuka M, Matsuo K, Ito Y, Yanagisawa A, Ohbiki M, Tabuchi K, Atsuta Y, Kanda J, Konuma T. The effect of center experience on allogeneic hematopoietic cell transplantation outcomes in acute myeloid leukemia. Bone Marrow Transplant 2024; 59:541-549. [PMID: 38321271 DOI: 10.1038/s41409-024-02222-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024]
Abstract
This study aimed to address the prognostic impact of center experience based on the data of 7821 adults with acute myeloid leukemia who underwent allogeneic hematopoietic cell transplantation (HCT) from 2010 to 2019 in Japan, where medical care was provided within a uniform healthcare system. Center experience was defined based on the number of allogeneic HCTs performed for any indication during the study period, by which centers were divided into low-, intermediate-, and high-volume centers. After adjusting for known confounding factors, the risk of overall mortality was lowest for the high-volume centers and highest for the low-volume centers, with the difference between the center categories attributed primarily to the risk of relapse. Patients transplanted at high-volume centers had higher risks of acute and chronic graft-versus-host diseases but without an increased risk of non-relapse mortality (NRM). These findings reveal the presence of a center effect in allogeneic HCT conducted during the past decade in Japan, highlighting the difference in relapse based on center experience. The weaker effect on NRM compared with that on relapse suggests that the transplantation care quality is becoming equalized across the country.
Collapse
Affiliation(s)
- Masamitsu Yanada
- Nagoya City University East Medical Center, Nagoya, Japan.
- Aichi Cancer Center, Nagoya, Japan.
| | - Shingo Yano
- The Jikei University School of Medicine, Tokyo, Japan
| | | | | | | | - Tatsuo Ichinohe
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | | | - Hideki Goto
- Hokkaido University Hospital, Sapporo, Japan
| | - Koji Kato
- Kyushu University Hospital, Fukuoka, Japan
| | - Fumihiko Ishimaru
- Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | | | | | | | - Yuri Ito
- Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Atsumi Yanagisawa
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
| | - Marie Ohbiki
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
- Aichi Medical University, Nagakute, Japan
| | - Ken Tabuchi
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
- Aichi Medical University, Nagakute, Japan
| | - Junya Kanda
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takaaki Konuma
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Zheng Y, Pan L, Li J, Feng X, Li C, Zheng M, Mai H, Yang L, He Y, He X, Xu H, Wen H, Le S. Prognostic significance of multiparametric flow cytometry minimal residual disease at two time points after induction in pediatric acute myeloid leukemia. BMC Cancer 2024; 24:46. [PMID: 38195455 PMCID: PMC10775489 DOI: 10.1186/s12885-023-11784-4] [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: 05/09/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Prompt response to induction chemotherapy is a prognostic factor in pediatric acute myeloid leukemia. In this study, we aimed to evaluate the prognostic significance of multiparametric flow cytometry-minimal residual disease (MFC-MRD), assessed at the end of the first and second induction courses. METHODS MFC-MRD was performed at the end of the first induction (TP1) in 524 patients and second induction (TP2) in 467 patients who were treated according to the modified Medical Research Council (UK) acute myeloid leukemia 15 protocol. RESULTS Using a 0.1% cutoff level, patients with MFC-MRD at the two time points had lower event-free survival and overall survival. Only the TP2 MFC-MRD level could predict the outcome in a separate analysis of high and intermediate risks based on European LeukemiaNet risk stratification and KMT2A rearrangement. The TP2 MFC-MRD level could further differentiate the prognosis of patients into complete remission or non-complete remission based on morphological evaluation. Multivariate analysis indicated the TP2 MFC-MRD level as an independent adverse prognostic factor for event-free survival and overall survival. When comparing patients with MFC-MRD ≥ 0.1%, those who underwent hematopoietic stem cell transplant during the first complete remission had significantly higher 5-year event-free survival and overall survival and lower cumulative incidence of relapse than those who only received consolidation chemotherapy. CONCLUSIONS The TP2 MFC-MRD level can predict the outcomes in pediatric patients with acute myeloid leukemia and help stratify post-remission treatment.
Collapse
Affiliation(s)
- Yongzhi Zheng
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lili Pan
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian Li
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoqin Feng
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunfu Li
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Mincui Zheng
- Department of Pediatric Hematology/Oncology, Hematology and Oncology, Hunan Children's Hospital, Changsha, China
| | - Huirong Mai
- Department of Pediatric Hematology/Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Lihua Yang
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yingyi He
- Department of Pediatric Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Xiangling He
- People's Hospital of Hunan Province, Changsha, China
| | - Honggui Xu
- Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Hong Wen
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Shaohua Le
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
11
|
Swaminathan M, Ravandi F. Can measurable residual disease assessment be reliably used to defer allogeneic stem cell transplant in patients with intermediate-risk acute myeloid leukemia? Haematologica 2023; 108:2561-2563. [PMID: 37345488 PMCID: PMC10543186 DOI: 10.3324/haematol.2023.283120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023] Open
Affiliation(s)
- Mahesh Swaminathan
- The University of Texas, MD Anderson Cancer Center, Department of Leukemia, Texas
| | - Farhad Ravandi
- The University of Texas, MD Anderson Cancer Center, Department of Leukemia, Texas.
| |
Collapse
|
12
|
Li X, Tong X. Role of Measurable Residual Disease in Older Adult Acute Myeloid Leukemia. Clin Interv Aging 2023; 18:921-931. [PMID: 37313310 PMCID: PMC10258117 DOI: 10.2147/cia.s409308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
There is overwhelming evidence indicating that the use of measurable residual disease (MRD) as a biomarker provides critical prognostic information and that MRD may have a role in directing postremission decisions. There are a variety of assays for MRD assessment, such as multiparameter flow cytometry and molecular assessment of MRD, which present different characteristics in patients older than 60 years of age. Due to multiple reasons related to age, the progress of older adult AML patients is rarely investigated, especially with respect to MRD. In this review, we will clarify the characteristics of different assays for assessing MRD, focusing on its role as a risk-stratification biomarker to predict prognostic information and its role in optimal postremission therapy among older adult AML patients. These characteristics also provide guidance regarding the potential to apply personalized medicine in older adult AML patients.
Collapse
Affiliation(s)
- Xueyao Li
- Department of Hematology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xiuzhen Tong
- Department of Hematology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
13
|
Srinivasan Rajsri K, Roy N, Chakraborty S. Acute Myeloid Leukemia Stem Cells in Minimal/Measurable Residual Disease Detection. Cancers (Basel) 2023; 15:2866. [PMID: 37345204 PMCID: PMC10216329 DOI: 10.3390/cancers15102866] [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: 04/06/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by an abundance of incompletely matured or immature clonally derived hematopoietic precursors called leukemic blasts. Rare leukemia stem cells (LSCs) that can self-renew as well as give rise to leukemic progenitors comprising the bulk of leukemic blasts are considered the cellular reservoir of disease initiation and maintenance. LSCs are widely thought to be relatively resistant as well as adaptive to chemotherapy and can cause disease relapse. Therefore, it is imperative to understand the molecular bases of LSC forms and functions during different stages of disease progression, so we can more accurately identify these cells and design therapies to target them. Irrespective of the morphological, cytogenetic, and cellular heterogeneity of AML, the uniform, singularly important and independently significant prognosticator of disease response to therapy and patient outcome is measurable or minimal residual disease (MRD) detection, defined by residual disease detection below the morphology-based 5% blast threshold. The importance of LSC identification and frequency estimation during MRD detection, in order to make MRD more effective in predicting disease relapse and modifying therapeutic regimen is becoming increasingly apparent. This review focuses on summarizing functional and cellular composition-based LSC identification and linking those studies to current techniques of MRD detection to suggest LSC-inclusive MRD detection as well as outline outstanding questions that need to be addressed to improve the future of AML clinical management and treatment outcomes.
Collapse
Affiliation(s)
- Kritika Srinivasan Rajsri
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; (K.S.R.); (N.R.)
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Nainita Roy
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; (K.S.R.); (N.R.)
| | - Sohini Chakraborty
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; (K.S.R.); (N.R.)
| |
Collapse
|
14
|
Ogbue O, Unlu S, Ibodeng GO, Singh A, Durmaz A, Visconte V, Molina JC. Single-Cell Next-Generation Sequencing to Monitor Hematopoietic Stem-Cell Transplantation: Current Applications and Future Perspectives. Cancers (Basel) 2023; 15:cancers15092477. [PMID: 37173944 PMCID: PMC10177286 DOI: 10.3390/cancers15092477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) are genetically complex and diverse diseases. Such complexity makes challenging the monitoring of response to treatment. Measurable residual disease (MRD) assessment is a powerful tool for monitoring response and guiding therapeutic interventions. This is accomplished through targeted next-generation sequencing (NGS), as well as polymerase chain reaction and multiparameter flow cytometry, to detect genomic aberrations at a previously challenging leukemic cell concentration. A major shortcoming of NGS techniques is the inability to discriminate nonleukemic clonal hematopoiesis. In addition, risk assessment and prognostication become more complicated after hematopoietic stem-cell transplantation (HSCT) due to genotypic drift. To address this, newer sequencing techniques have been developed, leading to more prospective and randomized clinical trials aiming to demonstrate the prognostic utility of single-cell next-generation sequencing in predicting patient outcomes following HSCT. This review discusses the use of single-cell DNA genomics in MRD assessment for AML/MDS, with an emphasis on the HSCT time period, including the challenges with current technologies. We also touch on the potential benefits of single-cell RNA sequencing and analysis of accessible chromatin, which generate high-dimensional data at the cellular resolution for investigational purposes, but not currently used in the clinical setting.
Collapse
Affiliation(s)
- Olisaemeka Ogbue
- Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH 44111, USA
| | - Serhan Unlu
- Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH 44111, USA
| | - Gogo-Ogute Ibodeng
- Internal Medicine, Infirmary Health's Thomas Hospital, Fairhope, AL 36607, USA
| | - Abhay Singh
- Department of Hematology Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Arda Durmaz
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Taussig Cancer Center, Cleveland, OH 44106, USA
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Taussig Cancer Center, Cleveland, OH 44106, USA
| | - John C Molina
- Department of Hematology Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH 44106, USA
| |
Collapse
|
15
|
Teixeira A, Carreira L, Abalde-Cela S, Sampaio-Marques B, Areias AC, Ludovico P, Diéguez L. Current and Emerging Techniques for Diagnosis and MRD Detection in AML: A Comprehensive Narrative Review. Cancers (Basel) 2023; 15:cancers15051362. [PMID: 36900154 PMCID: PMC10000116 DOI: 10.3390/cancers15051362] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Acute myeloid leukemia (AML) comprises a group of hematologic neoplasms characterized by abnormal differentiation and proliferation of myeloid progenitor cells. AML is associated with poor outcome due to the lack of efficient therapies and early diagnostic tools. The current gold standard diagnostic tools are based on bone marrow biopsy. These biopsies, apart from being very invasive, painful, and costly, have low sensitivity. Despite the progress uncovering the molecular pathogenesis of AML, the development of novel detection strategies is still poorly explored. This is particularly important for patients that check the criteria for complete remission after treatment, since they can relapse through the persistence of some leukemic stem cells. This condition, recently named as measurable residual disease (MRD), has severe consequences for disease progression. Hence, an early and accurate diagnosis of MRD would allow an appropriate therapy to be tailored, improving a patient's prognosis. Many novel techniques with high potential in disease prevention and early detection are being explored. Among them, microfluidics has flourished in recent years due to its ability at processing complex samples as well as its demonstrated capacity to isolate rare cells from biological fluids. In parallel, surface-enhanced Raman scattering (SERS) spectroscopy has shown outstanding sensitivity and capability for multiplex quantitative detection of disease biomarkers. Together, these technologies can allow early and cost-effective disease detection as well as contribute to monitoring the efficiency of treatments. In this review, we aim to provide a comprehensive overview of AML disease, the conventional techniques currently used for its diagnosis, classification (recently updated in September 2022), and treatment selection, and we also aim to present how novel technologies can be applied to improve the detection and monitoring of MRD.
Collapse
Affiliation(s)
- Alexandra Teixeira
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Luís Carreira
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
| | - Sara Abalde-Cela
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Anabela C. Areias
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (P.L.); (L.D.)
| | - Lorena Diéguez
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
- Correspondence: (P.L.); (L.D.)
| |
Collapse
|
16
|
Venugopal S, Xie Z, Zeidan AM. An overview of novel therapies in advanced clinical testing for acute myeloid leukemia. Expert Rev Hematol 2023; 16:109-119. [PMID: 36718500 DOI: 10.1080/17474086.2023.2174521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION The past decade has seen a sea change in the AML landscape with vastly improved cognizance of molecular pathogenesis, clonal evolution, and importance of measurable residual disease. Since 2017, the therapeutic armamentarium of AML has considerably expanded with the approval of midostaurin, enasidenib, ivosidenib, gilteritinib, and venetoclax in combination with hypomethylating agents and others. Nevertheless, relapse and treatment refractoriness remain the insurmountable challenges in AML therapy. This has galvanized the leukemic research community leading to the discovery and development of agents that specifically target gene mutations, molecularly agnostic therapies that exploit immune environment, apoptotic pathways, leukemic cell surface antigens and so forth. AREAS COVERED This article provides an overview of the pathophysiology of AML in the context of non-cellular immune and molecularly targeted and agnostic therapies that are in clinical trial development in AML. EXPERT OPINION Ever growing understanding of the molecular pathogenesis and metabolomics in AML has allowed the researchers to identify targets directed at specific genes and metabolic pathways. As a result, AML therapy is constantly evolving and so are the escape mechanisms leading to disease relapse. Therefore, it is of paramount importance to sequentially evaluate the patient during AML treatment and intervene at the right time.
Collapse
Affiliation(s)
- Sangeetha Venugopal
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Zhuoer Xie
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, FL, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
17
|
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
|
18
|
Azenkot T, Jonas BA. Clinical Impact of Measurable Residual Disease in Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:cancers14153634. [PMID: 35892893 PMCID: PMC9330895 DOI: 10.3390/cancers14153634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Advances in immunophenotyping and molecular techniques have allowed for the development of more sensitive diagnostic tests in acute leukemia. These techniques can identify low levels of leukemic cells (quantified as 10−4 to 10−6 ratio to white blood cells) in patient samples. The presence of such low levels of leukemic cells, termed “measurable/minimal residual disease” (MRD), has been shown to be a marker of disease burden and patient outcomes. In acute lymphoblastic leukemia, new agents are highly effective at eliminating MRD for patients whose leukemia progressed despite first line therapies. By comparison, the role of MRD in acute myeloid leukemia is less clear. This commentary reviews select data and remaining questions about the clinical application of MRD to the treatment of patients with acute myeloid leukemia. Abstract Measurable residual disease (MRD) has emerged as a primary marker of risk severity and prognosis in acute myeloid leukemia (AML). There is, however, ongoing debate about MRD-based surveillance and treatment. A literature review was performed using the PubMed database with the keywords MRD or residual disease in recently published journals. Identified articles describe the prognostic value of pre-transplant MRD and suggest optimal timing and techniques to quantify MRD. Several studies address the implications of MRD on treatment selection and hematopoietic stem cell transplant, including patient candidacy, conditioning regimen, and transplant type. More prospective, randomized studies are needed to guide the application of MRD in the treatment of AML, particularly in transplant.
Collapse
Affiliation(s)
- Tali Azenkot
- Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| | - Brian A. Jonas
- Division of Cellular Therapy, Bone Marrow Transplant, and Malignant Hematology, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Correspondence: ; Tel.: +1-916-734-3772
| |
Collapse
|
19
|
Virk H, Khaire N, Sreedharanunni S, Naseem S, Malhotra P, Sharma P. Significance of Dysplastic Neutrophils with Multiple Auer Rods in Post-therapy Acute Promyelocytic Leukemia. Indian J Hematol Blood Transfus 2022; 38:199-201. [PMID: 35125730 PMCID: PMC8804073 DOI: 10.1007/s12288-021-01493-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/21/2021] [Indexed: 01/03/2023] Open
Affiliation(s)
- Harpreet Virk
- Department of Hematology, Postgraduate Institute of Medical Education & Research, Level 5, Research Block A Sector 12, Chandigarh, 160012 India
| | - Niranjan Khaire
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Sreejesh Sreedharanunni
- Department of Hematology, Postgraduate Institute of Medical Education & Research, Level 5, Research Block A Sector 12, Chandigarh, 160012 India
| | - Shano Naseem
- Department of Hematology, Postgraduate Institute of Medical Education & Research, Level 5, Research Block A Sector 12, Chandigarh, 160012 India
| | - Pankaj Malhotra
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Prashant Sharma
- Department of Hematology, Postgraduate Institute of Medical Education & Research, Level 5, Research Block A Sector 12, Chandigarh, 160012 India
| |
Collapse
|
20
|
Récher C. The beginning of a new therapeutic era in acute myeloid leukemia. EJHAEM 2021; 2:823-833. [PMID: 35845213 PMCID: PMC9175720 DOI: 10.1002/jha2.252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/17/2022]
Abstract
In the field of AML, the early 2000s were shaped by the advent of novel molecular biology technologies including high-throughput sequencing that improved prognostic classification, response evaluation through the quantification of minimal residual disease, and the launch of research on targeted therapies. Our knowledge of leukemogenesis, AML genetic diversity, gene-gene interactions, clonal evolution, and treatment response assessment has also greatly improved. New classifications based on chromosomal abnormalities and gene mutations are now integrated on a routine basis. These considerable efforts contributed to the discovery and development of promising drugs which specifically target gene mutations, apoptotic pathways and cell surface antigens as well as reformulate classical cytotoxic agents. In less than 2 years, nine novels drugs have been approved for the treatment of AML patients, and many others are being intensively investigated, in particular immune therapies. There are now numerous clinical research opportunities offered to clinicians, thanks to these new treatment options. We are only at the start of a new era which should see major disruptions in the way we understand, treat, and monitor patients with AML.
Collapse
Affiliation(s)
- Christian Récher
- Service d'HématologieCentre Hospitalier Universitaire de ToulouseInstitut Universitaire du Cancer de Toulouse OncopoleUniversité Toulouse III Paul SabatierCentre de Recherches en Cancérologie de ToulouseToulouseFrance
| |
Collapse
|
21
|
Dinh KN, Jaksik R, Corey SJ, Kimmel M. Predicting Time to Relapse in Acute Myeloid Leukemia through Stochastic Modeling of Minimal Residual Disease Based on Clonality Data. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2021; 1. [PMID: 34541576 DOI: 10.1002/cso2.1026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Event-free and overall survival remain poor for patients with acute myeloid leukemia. Chemoresistant clones contributing to relapse arise from minimal residual disease (MRD) or newly-acquired mutations. However, the dynamics of clones comprising MRD is poorly understood. We developed a predictive stochastic model, based on a multitype age-dependent Markov branching process, to describe how random events in MRD contribute to the heterogeneity in treatment response. We employed training and validation sets of patients who underwent whole genome sequencing and for whom mutant clone frequencies at diagnosis and relapse were available. The disease evolution and treatment outcome are subject to stochastic fluctuations. Estimates of malignant clone growth rates, obtained by model fitting, are consistent with published data. Using the estimates from the training set, we developed a function linking MRD and time of relapse, with MRD inferred from the model fits to clone frequencies and other data. An independent validation set confirmed our model. In a third data set, we fitted the model to data at diagnosis and remission and predicted the time to relapse. As a conclusion, given bone marrow genome at diagnosis and MRD at or past remission, the model can predict time to relapse, and help guide treatment decisions to mitigate relapse.
Collapse
Affiliation(s)
- Khanh N Dinh
- Irving Institute of Cancer Dynamics, Columbia University, New York, NY, USA
| | - Roman Jaksik
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Seth J Corey
- Departments of Pediatric Hematology/Oncology and Stem Cell Transplantation and Cancer Biology, Cleveland Clinic, Cleveland, OH, USA
| | - Marek Kimmel
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland.,Department of Statistics, Rice University, Houston, TX, USA
| |
Collapse
|
22
|
Aitken MJL, Ravandi F, Patel KP, Short NJ. Prognostic and therapeutic implications of measurable residual disease in acute myeloid leukemia. J Hematol Oncol 2021; 14:137. [PMID: 34479626 PMCID: PMC8417965 DOI: 10.1186/s13045-021-01148-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/25/2021] [Indexed: 01/10/2023] Open
Abstract
Quantification of measurable residual disease (MRD) provides critical prognostic information in acute myeloid leukemia (AML). A variety of platforms exist for MRD detection, varying in their sensitivity and applicability to individual patients. MRD detected by quantitative polymerase chain reaction, multiparameter flow cytometry, or next-generation sequencing has prognostic implications in various subsets of AML and at various times throughout treatment. While it is overwhelmingly evident that minute levels of remnant disease confer increased risk of relapse and shortened survival, the therapeutic implications of MRD remain less clear. The use of MRD as a guide to selecting the most optimal post-remission therapy, including hematopoietic stem cell transplant or maintenance therapy with hypomethylating agents, small molecule inhibitors, or immunotherapy is an area of active investigation. In addition, whether there are sufficient data to use MRD negativity as a surrogate endpoint in clinical trial development is controversial. In this review, we will critically examine the methods used to detect MRD, its role as a prognostic biomarker, MRD-directed therapeutics, and its potential role as a study endpoint.
Collapse
Affiliation(s)
- Marisa J L Aitken
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,McGovern Medical School, UT Health Science Center-Houston, Houston, TX, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Maiti A, DiNardo CD, Qiao W, Kadia TM, Jabbour EJ, Rausch CR, Daver NG, Short NJ, Borthakur G, Pemmaraju N, Yilmaz M, Alvarado Y, Montalbano KS, Wade A, Maduike RE, Guerrero JA, Vaughan K, Bivins CA, Pierce S, Ning J, Ravandi F, Kantarjian HM, Konopleva MY. Ten-day decitabine with venetoclax versus intensive chemotherapy in relapsed or refractory acute myeloid leukemia: A propensity score-matched analysis. Cancer 2021; 127:4213-4220. [PMID: 34343352 DOI: 10.1002/cncr.33814] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 06/04/2021] [Accepted: 06/23/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Relapsed/refractory (R/R) acute myeloid leukemia (AML) has poor outcomes. Although lower-intensity venetoclax-containing regimens are standard for older/unfit patients with newly diagnosed AML, it is unknown how such regimens compare with intensive chemotherapy (IC) for R/R AML. METHODS Outcomes of R/R AML treated with 10-day decitabine and venetoclax (DEC10-VEN) were compared with IC-based regimens including idarubicin with cytarabine, with or without cladribine, clofarabine, or fludarabine, with or without additional agents. Propensity scores derived from patient baseline characteristics were used to match DEC10-VEN and IC patients to minimize bias. RESULTS Sixty-five patients in the DEC10-VEN cohort were matched to 130 IC recipients. The median ages for the DEC10-VEN and IC groups were 64 and 58 years, respectively, and baseline characteristics were balanced between the 2 cohorts. DEC10-VEN conferred significantly higher responses compared with IC including higher overall response rate (60% vs 36%; odds ratio [OR], 3.28; P < .001), complete remission with incomplete hematologic recovery (CRi, 19% vs 6%; OR, 3.56; P = .012), minimal residual disease negativity by flow cytometry (28% vs 13%; OR, 2.48; P = .017), and lower rates of refractory disease. DEC10-VEN led to significantly longer median event-free survival compared with IC (5.7 vs 1.5 months; hazard ratio [HR], 0.46; 95% CI, 0.30-0.70; P < .001), as well as median overall survival (OS; 6.8 vs 4.7 months; HR, 0.56; 95% CI, 0.37-0.86; P = .008). DEC10-VEN was independently associated with improved OS compared with IC in multivariate analysis. Exploratory analysis for OS in 27 subgroups showed that DEC10-VEN was comparable with IC as salvage therapy for R/R AML. CONCLUSION DEC10-VEN represents an appropriate salvage therapy and may offer better responses and survival compared with IC in adults with R/R AML.
Collapse
Affiliation(s)
- Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Qiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elias J Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Caitlin R Rausch
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yesid Alvarado
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathryn S Montalbano
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Allison Wade
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rita E Maduike
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julio A Guerrero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kenneth Vaughan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carol A Bivins
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Y Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
24
|
Talami A, Bettelli F, Pioli V, Giusti D, Gilioli A, Colasante C, Galassi L, Giubbolini R, Catellani H, Donatelli F, Maffei R, Martinelli S, Barozzi P, Potenza L, Marasca R, Trenti T, Tagliafico E, Comoli P, Luppi M, Forghieri F. How to Improve Prognostication in Acute Myeloid Leukemia with CBFB-MYH11 Fusion Transcript: Focus on the Role of Molecular Measurable Residual Disease (MRD) Monitoring. Biomedicines 2021; 9:biomedicines9080953. [PMID: 34440157 PMCID: PMC8391269 DOI: 10.3390/biomedicines9080953] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) carrying inv(16)/t(16;16), resulting in fusion transcript CBFB-MYH11, belongs to the favorable-risk category. However, even if most patients obtain morphological complete remission after induction, approximately 30% of cases eventually relapse. While well-established clinical features and concomitant cytogenetic/molecular lesions have been recognized to be relevant to predict prognosis at disease onset, the independent prognostic impact of measurable residual disease (MRD) monitoring by quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR), mainly in predicting relapse, actually supersedes other prognostic factors. Although the ELN Working Party recently indicated that patients affected with CBFB-MYH11 AML should have MRD assessment at informative clinical timepoints, at least after two cycles of intensive chemotherapy and after the end of treatment, several controversies could be raised, especially on the frequency of subsequent serial monitoring, the most significant MRD thresholds (most commonly 0.1%) and on the best source to be analyzed, namely, bone marrow or peripheral blood samples. Moreover, persisting low-level MRD positivity at the end of treatment is relatively common and not predictive of relapse, provided that transcript levels remain stably below specific thresholds. Rising MRD levels suggestive of molecular relapse/progression should thus be confirmed in subsequent samples. Further prospective studies would be required to optimize post-remission monitoring and to define effective MRD-based therapeutic strategies.
Collapse
Affiliation(s)
- Annalisa Talami
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Francesca Bettelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Valeria Pioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Davide Giusti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Andrea Gilioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Corrado Colasante
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Laura Galassi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Rachele Giubbolini
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Hillary Catellani
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Francesca Donatelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Rossana Maffei
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Silvia Martinelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Patrizia Barozzi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Leonardo Potenza
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Roberto Marasca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Unità Sanitaria Locale, 41126 Modena, Italy;
| | - Enrico Tagliafico
- Center for Genome Research, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy;
| | - Patrizia Comoli
- Pediatric Hematology/Oncology Unit and Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, 27100 Pavia, Italy;
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
- Correspondence: (M.L.); (F.F.); Tel.: +39-059-4222447 (F.F.); Fax: +39-059-4222386 (F.F.)
| | - Fabio Forghieri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
- Correspondence: (M.L.); (F.F.); Tel.: +39-059-4222447 (F.F.); Fax: +39-059-4222386 (F.F.)
| |
Collapse
|
25
|
Selove W, Hutchinson L, Makarenko V, Meng X, Tomaszewicz K, Ramanathan M, Cerny J, Nath R, Chen B, Woda B, Bledsoe JR. Impact of pretransplant mutation status on survival after allogeneic stem cell transplant for acute myeloid leukemia. EJHAEM 2021; 2:514-519. [PMID: 35844698 PMCID: PMC9175694 DOI: 10.1002/jha2.260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 11/24/2022]
Affiliation(s)
- William Selove
- Department of PathologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| | - Lloyd Hutchinson
- Department of PathologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| | - Vladislav Makarenko
- Department of PathologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| | - Xiuling Meng
- Department of PathologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| | - Keith Tomaszewicz
- Department of PathologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| | - Muthalagu Ramanathan
- Department of Hematology‐OncologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| | - Jan Cerny
- Department of Hematology‐OncologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| | - Rajneesh Nath
- Department of HematologyMedical OncologyBanner MD Anderson Cancer Center ClinicGilbertArizonaUSA
| | - Benjamin Chen
- Bristol Myers Squibb CompanyCambridgeMassachusettsUSA
| | - Bruce Woda
- Department of PathologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| | - Jacob R. Bledsoe
- Department of PathologyUMass Memorial Medical CenterUniversity of MassachusettsWorcesterMassachusettsUSA
| |
Collapse
|
26
|
Yu S, Fan Z, Ma L, Wang Y, Huang F, Zhang Q, Huang J, Wang S, Xu N, Xuan L, Xiong M, Han L, Sun Z, Zhang H, Liu H, Yu G, Shi P, Xu J, Wu M, Guo Z, Xiong Y, Duan C, Sun J, Liu Q, Zhang Y. Association Between Measurable Residual Disease in Patients With Intermediate-Risk Acute Myeloid Leukemia and First Remission, Treatment, and Outcomes. JAMA Netw Open 2021; 4:e2115991. [PMID: 34232303 PMCID: PMC8264648 DOI: 10.1001/jamanetworkopen.2021.15991] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/03/2021] [Indexed: 12/30/2022] Open
Abstract
Importance Measurable residual disease (MRD) is widely used as a therapy-stratification factor for acute myeloid leukemia (AML), but the association of dynamic MRD with postremission treatment (PRT) in patients with intermediate-risk AML (IR-AML) has not been well investigated. Objective To investigate PRT choices based on dynamic MRD in patients with IR-AML. Design, Setting, and Participants This cohort study examined 549 younger patients with de novo IR-AML in the South China Hematology Alliance database during the period from January 1, 2012, to June 30, 2016, including 154 who received chemotherapy, 116 who received an autologous stem cell transplant (auto-SCT), and 279 who received an allogeneic SCT (allo-SCT). Subgroup analyses were performed according to dynamic MRD after the first, second, and third courses of chemotherapy. The end point of the last follow-up was August 31, 2020. Statistical analysis was performed from December 1, 2019, to September 30, 2020. Exposures Receipt of chemotherapy, auto-SCT, or allo-SCT. Main Outcomes and Measures The primary end points were 5-year cumulative incidence of relapse and leukemia-free survival. Results Subgroup analyses were performed for 549 participants (314 male participants [57.2%]; median age, 37 years [range, 14-60 years]) according to the dynamics of MRD after 1, 2, or 3 courses of chemotherapy. Comparable cumulative incidences of relapse, leukemia-free survival, and overall survival were observed among participants who had no MRD after 1, 2, or 3 courses of chemotherapy. Participants who underwent chemotherapy and those who underwent auto-SCT had better graft-vs-host disease-free, relapse-free survival (GRFS) than those who underwent allo-SCT (chemotherapy: hazard ratio [HR], 0.35 [95% CI, 0.14-0.90]; P = .03; auto-SCT: HR, 0.07 [95% CI, 0.01-0.58]; P = .01). Among participants with MRD after 1 course of chemotherapy but no MRD after 2 or 3 courses, those who underwent auto-SCT and allo-SCT showed lower cumulative incidence of relapse (auto-SCT: HR, 0.25 [95% CI, 0.08-0.78]; P = .01; allo-SCT: HR, 0.08 [95% CI, 0.02-0.24]; P < .001), better leukemia-free survival (auto-SCT: HR, 0.26 [95% CI, 0.10-0.64]; P = .004; allo-SCT: HR, 0.21 [95% CI, 0.09-0.46]; P < .001), and overall survival (auto-SCT: HR, 0.22 [95% CI, 0.08-0.64]; P = .005; allo-SCT: HR, 0.25 [95% CI, 0.11-0.59]; P = .001) vs chemotherapy. In addition, auto-SCT showed better GRFS than allo-SCT (HR, 0.45 [95% CI, 0.21-0.98]; P = .04) in this group. Among participants with MRD after 1 or 2 courses of chemotherapy but no MRD after 3 courses, allo-SCT had superior cumulative incidence of relapse (HR, 0.10 [95% CI, 0.06-0.94]; P = .04) and leukemia-free survival (HR, 0.18 [95% CI, 0.05-0.68]; P = .01) compared with chemotherapy, but no advantageous cumulative incidence of relapse (HR, 0.15 [95% CI, 0.02-1.42]; P = .10) and leukemia-free survival (HR, 0.23 [95% CI, 0.05-1.08]; P = .06) compared with auto-SCT. Among participants with MRD after 3 courses of chemotherapy, allo-SCT had superior cumulative incidences of relapse, leukemia-free survival, and overall survival compared with chemotherapy (relapse: HR, 0.16 [95% CI, 0.08-0.33]; P < .001; leukemia-free survival: HR, 0.19 [95% CI, 0.10-0.35]; P < .001; overall survival: HR, 0.29 [95% CI, 0.15-0.55]; P < .001) and auto-SCT (relapse: HR, 0.25 [95% CI, 0.12-0.53]; P < .001; leukemia-free survival: HR, 0.35 [95% CI, 0.18-0.73]; P = .004; overall survival: HR, 0.54 [95% CI, 0.26-0.94]; P = .04). Among participants with recurrent MRD, allo-SCT was also associated with advantageous cumulative incidence of relapse, leukemia-free survival, and overall survival compared with chemotherapy (relapse: HR, 0.12 [95% CI, 0.04-0.33]; P < .001; leukemia-free survival: HR, 0.24 [95% CI, 0.10-0.56]; P = .001; overall survival: HR, 0.31 [95% CI, 0.13-0.75]; P = .01) and auto-SCT (relapse: HR, 0.28 [95% CI, 0.09-0.81]; P = .02; leukemia-free survival: HR, 0.30 [95% CI, 0.12-0.76]; P = .01; overall survival: HR, 0.26 [95% CI, 0.10-0.70]; P = .007). Conclusions and Relevance This study suggests that clinical decisions based on dynamic MRD might be associated with improved therapy stratification and optimized PRT for patients with IR-AML. Prospective multicenter trials are needed to further validate these findings.
Collapse
Affiliation(s)
- Sijian Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiping Fan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liping Ma
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Yu Wang
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China
| | - Fen Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qing Zhang
- Department of Hematology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jiafu Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People’s Hospital, Guangzhou, China
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mujun Xiong
- Department of Hematology, The First People’s Hospital of Chenzhou, Chenzhou, China
| | - Lijie Han
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiqiang Sun
- Department of Hematology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Hongyu Zhang
- Department of Hematology, Shenzhen Hospital of Peking University, Shenzhen, China
| | - Hui Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guopan Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengcheng Shi
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meiqing Wu
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Ziwen Guo
- Department of Hematology, Zhongshan People’s Hospital, Zhongshan, China
| | - Yiying Xiong
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chongyang Duan
- Department of Biostatistics, Southern Medical University School of Public Health, Guangzhou, China
| | - Jing Sun
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Bernasconi P, Borsani O. Eradication of Measurable Residual Disease in AML: A Challenging Clinical Goal. Cancers (Basel) 2021; 13:3170. [PMID: 34202000 PMCID: PMC8268140 DOI: 10.3390/cancers13133170] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 12/18/2022] Open
Abstract
In non-promyelocytic (non-M3) AML measurable residual disease (MRD) detected by multi-parameter flow cytometry and molecular technologies, which are guided by Consensus-based guidelines and discover very low leukemic cell numbers far below the 5% threshold of morphological assessment, has emerged as the most relevant predictor of clinical outcome. Currently, it is well-established that MRD positivity after standard induction and consolidation chemotherapy, as well as during the period preceding an allogeneic hematopoietic stem cell transplant (allo-HSCT), portends to a significantly inferior relapse-free survival (RFS) and overall survival (OS). In addition, it has become absolutely clear that conversion from an MRD-positive to an MRD-negative state provides a favorable clinical outcome similar to that associated with early MRD negativity. Thus, the complete eradication of MRD, i.e., the clearance of the few leukemic stem cells-which, due to their chemo-radiotherapy resistance, might eventually be responsible of disease recurrence-has become an un-met clinical need in AML. Nowadays, this goal might potentially be achieved thanks to the development of novel innovative treatment strategies, including those targeting driver mutations, apoptosis, methylation patterns and leukemic proteins. The aim of this review is to analyze these strategies and to suggest any potential combination able to induce MRD negativity in the pre- and post-HSCT period.
Collapse
Affiliation(s)
- Paolo Bernasconi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
- Hematology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Oscar Borsani
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
28
|
The evolving concept of indications for allogeneic hematopoietic cell transplantation during first complete remission of acute myeloid leukemia. Bone Marrow Transplant 2021; 56:1257-1265. [PMID: 33686251 DOI: 10.1038/s41409-021-01247-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 02/08/2023]
Abstract
The long-standing debate of whether patients with acute myeloid leukemia (AML) should proceed to allogeneic hematopoietic cell transplantation (HCT) during first complete remission (CR1) remains unsettled. Although allogeneic HCT during CR1 used to be recommended for those with intermediate or poor cytogenetics if they had a matched sibling donor, the concept of indications for allogeneic HCT during CR1 has been evolving by virtue of advances in understanding of the molecular pathogenesis of AML and innovations in transplantation practice attained over the last few decades. The incorporation of molecular profiles of leukemia has been shown to contribute to further refinements of risk classification that had previously relied mostly on cytogenetics, while the progress in transplantation procedures has made it possible to perform transplantations more safely even for patients without a matched sibling donor. These significant changes have underpinned the need to reappraise indications for allogeneic HCT during CR1 of AML. Improvements in clinical applications of genetic and measurable residual disease information as well as in transplantation technology are expected to further refine indications for allogeneic HCT during CR1, and thus promote an individualized approach for the treatment of AML.
Collapse
|
29
|
Prognostic value of measurable residual disease after venetoclax and decitabine in acute myeloid leukemia. Blood Adv 2021; 5:1876-1883. [PMID: 33792630 DOI: 10.1182/bloodadvances.2020003717] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/11/2021] [Indexed: 12/21/2022] Open
Abstract
Assessment of measurable residual disease (MRD) provides prognostic information in acute myeloid leukemia (AML). However, the utility of MRD with venetoclax-based lower intensity regimens is unknown. We analyzed the prognostic value of achieving a negative MRD in older/"unfit" patients with AML receiving first-line therapy with 10-day decitabine and venetoclax. MRD was evaluated in bone marrow specimens using multicolor flow cytometry (sensitivity 0.1%). Ninety-seven patients achieving either a complete remission (CR) or CR with incomplete hematologic recovery (CRi) or morphologic leukemia-free state were included. Median age was 72 years (interquartile range, 68-78 years), and 64% had adverse-risk AML. Eighty-three patients achieved CR/CRi, and 52 (54%) became MRD negative. Median time to becoming MRD negative was 2.0 months (interquartile range, 0.9-3.1 months). Patients becoming MRD negative by 2 months had longer relapse-free survival (RFS) compared with those remaining MRD positive (median RFS, not reached vs 5.2 months; hazard ratio [HR], 0.31; 95% confidence interval [CI], 0.12-0.78; P = .004), longer event-free survival (EFS) (median EFS, not reached vs 5.8 months; HR, 0.25; 95% CI, 0.12-0.55; P < .001), as well as longer overall survival (OS) (median OS, 25.1 vs 7.1 months; HR, 0.23; 95% CI, 0.11-0.51; P < .001). Patients achieving an MRD-negative CR had longer OS compared with those with an inferior response (median OS, 25.1 vs 11.6 months; HR, 0.33; 95% CI, 0.19-0.58; P < .0005). Patients becoming MRD negative within 1 month had an improved OS compared with MRD-positive patients (median OS, 25.1 vs 3.4 months; HR, 0.15; 95% CI, 0.03-0.64; P < .0001). Differential impact of MRD status on survival outcomes persisted at a later 4-month time point of evaluation. In conclusion, MRD-negative status at 1, 2, and 4 months after starting therapy confers significantly better survival in older/unfit patients with AML receiving first-line therapy with 10-day decitabine and venetoclax. This trial was registered at www.clinicaltrials.gov as #NCT03404193.
Collapse
|
30
|
Othman TA, Azenkot T, Moskoff BN, Tenold ME, Jonas BA. Venetoclax-based combinations for the treatment of newly diagnosed acute myeloid leukemia. Future Oncol 2021; 17:2989-3005. [PMID: 34024158 DOI: 10.2217/fon-2021-0262] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Elderly and/or unfit patients with acute myeloid leukemia have historically been challenging to manage as they were ineligible for what was considered standard of care treatment with induction chemotherapy. The emergence of venetoclax with hypomethylating agents or low-dose cytarabine has substantially improved outcomes in the frontline setting with manageable toxicity. However, this regimen can be challenging to deliver given its differences from standard intensive chemotherapy. In this review, we summarize the landmark trials that established venetoclax-based combinations as a new standard of care for patients with acute myeloid leukemia not suitable for intense chemotherapy, provide practical clinical pearls for managing patients on these therapies, and offer a brief overview of modifications to these regimens under development to improve their efficacy and/or applicability.
Collapse
Affiliation(s)
- Tamer A Othman
- Department of Internal Medicine, Division of Hematology & Oncology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Tali Azenkot
- Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Benjamin N Moskoff
- Pharmacy Department, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Matthew E Tenold
- Department of Internal Medicine, Division of Hematology & Oncology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Brian A Jonas
- Department of Internal Medicine, Division of Hematology & Oncology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
31
|
Prognostic impact of complete remission with MRD negativity in patients with relapsed or refractory AML. Blood Adv 2021; 4:6117-6126. [PMID: 33351107 DOI: 10.1182/bloodadvances.2020002811] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
In relapsed/refractory acute myeloid leukemia (AML), the prognostic impact of complete remission (CR) and measurable residual disease (MRD) negativity is not well established. We retrospectively analyzed 141 patients with relapsed/refractory AML who received first salvage therapy and had MRD assessed by multiparameter flow cytometry at the time of response. Patients who achieved CR with full hematologic recovery as best response vs those with incomplete hematology recovery had lower cumulative incidence of relapse (P = .01) and better relapse-free survival (P = .004) but not overall survival (P = .15); a similar trend was observed in patients who achieved MRD negativity vs those who were MRD positive (P = .01, P = .05, and P = .21, respectively). By multivariate analysis, CR and MRD negativity were each independently associated with lower cumulative incidence of relapse (P = .001 and P = .003, respectively) and better relapse-free survival (P < .001 and P = .02) but not overall survival. Patients who achieved CR with MRD negativity had the lowest rates of relapse and best survival (2-year overall survival rate, 37%), which was driven largely by lower rates of early relapse and an increased ability in this group to undergo hematopoietic stem cell transplantation (HSCT); however, post-HSCT outcomes were similar regardless of response to salvage chemotherapy. Overall, in patients with relapsed/refractory AML, CR with MRD negativity was associated with the best outcomes, supporting it as the optimal response in this setting.
Collapse
|
32
|
Reikvam H, Johansen S, Koenecke C. Future perspective: precision medicine to improve treatment results in the settings of allogenic stem cell transplantation for acute myelogenous leukemia. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1897464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Håkon Reikvam
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Silje Johansen
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
33
|
Trino S, Lamorte D, Caivano A, De Luca L, Sgambato A, Laurenzana I. Clinical relevance of extracellular vesicles in hematological neoplasms: from liquid biopsy to cell biopsy. Leukemia 2021; 35:661-678. [PMID: 33299143 PMCID: PMC7932927 DOI: 10.1038/s41375-020-01104-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/30/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
In the era of precision medicine, liquid biopsy is becoming increasingly important in oncology. It consists in the isolation and analysis of tumor-derived biomarkers, including extracellular vesicles (EVs), in body fluids. EVs are lipid bilayer-enclosed particles, heterogeneous in size and molecular composition, released from both normal and neoplastic cells. In tumor context, EVs are valuable carriers of cancer information; in fact, their amount, phenotype and molecular cargo, including proteins, lipids, metabolites and nucleic acids, mirror nature and origin of parental cells rendering EVs appealing candidates as novel biomarkers. Translation of these new potential diagnostic tools into clinical practice could deeply revolutionize the cancer field mainly for solid tumors but for hematological neoplasms, too.
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy.
| | - Antonella Caivano
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Luciana De Luca
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Alessandro Sgambato
- Scientific Direction, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy.
| |
Collapse
|
34
|
Short NJ, Zhou S, Fu C, Berry DA, Walter RB, Freeman SD, Hourigan CS, Huang X, Nogueras Gonzalez G, Hwang H, Qi X, Kantarjian H, Ravandi F. Association of Measurable Residual Disease With Survival Outcomes in Patients With Acute Myeloid Leukemia: A Systematic Review and Meta-analysis. JAMA Oncol 2020; 6:1890-1899. [PMID: 33030517 PMCID: PMC7545346 DOI: 10.1001/jamaoncol.2020.4600] [Citation(s) in RCA: 241] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022]
Abstract
IMPORTANCE Measurable residual disease (MRD) refers to neoplastic cells that cannot be detected by standard cytomorphologic analysis. In patients with acute myeloid leukemia (AML), determining the association of MRD with survival may improve prognostication and inform selection of efficient clinical trial end points. OBJECTIVE To examine the association between MRD status and disease-free survival (DFS) and overall survival (OS) in patients with AML using scientific literature. DATA SOURCES Clinical studies on AML published between January 1, 2000, and October 1, 2018, were identified via searches of PubMed, Embase, and MEDLINE. STUDY SELECTION Literature search and study screening were performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. Studies that assessed DFS or OS by MRD status in patients with AML were included. Reviews, non-English-language articles, and studies reporting only outcomes after hematopoietic cell transplantation or those with insufficient description of MRD information were excluded. DATA EXTRACTION AND SYNTHESIS Study sample size, median patient age, median follow-up time, MRD detection method, MRD assessment time points, AML subtype, specimen source, and survival outcomes were extracted. Meta-analyses were performed separately for DFS and OS using bayesian hierarchical modeling. MAIN OUTCOMES AND MEASURES Meta-analyses of survival probabilities and hazard ratios (HRs) were conducted for OS and DFS according to MRD status. RESULTS Eighty-one publications reporting on 11 151 patients were included. The average HR for achieving MRD negativity was 0.36 (95% bayesian credible interval [CrI], 0.33-0.39) for OS and 0.37 (95% CrI, 0.34-0.40) for DFS. The estimated 5-year DFS was 64% for patients without MRD and 25% for those with MRD, and the estimated OS was 68% for patients without MRD and 34% for those with MRD. The association of MRD negativity with DFS and OS was significant for all subgroups, with the exception of MRD assessed by cytogenetics or fluorescent in situ hybridization. CONCLUSIONS AND RELEVANCE The findings of this meta-analysis suggest that achievement of MRD negativity is associated with superior DFS and OS in patients with AML. The value of MRD negativity appears to be consistent across age groups, AML subtypes, time of MRD assessment, specimen source, and MRD detection methods. These results support MRD status as an end point that may allow for accelerated evaluation of novel therapies in AML.
Collapse
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Shouhao Zhou
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Chenqi Fu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Donald A. Berry
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sylvie D. Freeman
- Institute of Infection and Immunity, University of Birmingham, Birmingham, United Kingdom
| | - Christopher S. Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | | | - Hyunsoo Hwang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Xinyue Qi
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
35
|
Rotchanapanya W, Hokland P, Tunsing P, Owattanapanich W. Clinical Outcomes Based on Measurable Residual Disease Status in Patients with Core-Binding Factor Acute Myeloid Leukemia: A Systematic Review and Meta-Analysis. J Pers Med 2020; 10:jpm10040250. [PMID: 33256157 PMCID: PMC7711894 DOI: 10.3390/jpm10040250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 01/11/2023] Open
Abstract
Measurable residual disease (MRD) response during acute myeloid leukemia (AML) treatment is a gold standard for determining treatment strategy, especially in core-binding factor (CBL) AML. The aim of this study was to critically review the literature on MRD status in the CBF-AML to determine the overall impact of MRD status on clinical outcomes. Published studies in the MEDLINE and EMBASE databases from their inception up to 1 June 2019 were searched. The primary end-point was either overall survival (OS) or recurrence-free survival (RFS) between MRD negative and MRD positive CBF-AML patients. The secondary variable was cumulative incidence of relapse (CIR) between groups. Of the 736 articles, 13 relevant studies were included in this meta-analysis. The MRD negative group displayed more favorable recurrence-free survival (RFS) than those with MRD positivity, with a pooled odds ratio (OR) of 4.5. Moreover, OS was also superior in the MRD negative group, with a pooled OR of 7.88. Corroborating this, the CIR was statistically significantly lower in the MRD negative group, with a pooled OR of 0.06. The most common cutoff MRD level was 1 × 10−3. These results suggest that MRD assessment should be a routine investigation in clinical practice in this AML subset.
Collapse
Affiliation(s)
- Wannaphorn Rotchanapanya
- Division of Hematology, Department of Medicine, Chiangrai Prachanukroh Hospital, Chiang Rai 57000, Thailand;
| | - Peter Hokland
- Division of Hematology, Department of Clinical Medicine, Aarhus University Hospital, 8200 Aarhus N, Denmark;
| | - Pattaraporn Tunsing
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Weerapat Owattanapanich
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
- Correspondence: ; Tel.: +66-2419-4448
| |
Collapse
|
36
|
Gaut D, Mead M. Measurable residual disease in hematopoietic stem cell transplantation-eligible patients with acute myeloid leukemia: clinical significance and promising therapeutic strategies. Leuk Lymphoma 2020; 62:8-31. [DOI: 10.1080/10428194.2020.1827251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Daria Gaut
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Monica Mead
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
37
|
de Melo Rodrigues AL, Bonfim C, Seber A, Colturato VAR, Zecchin VG, Nichele S, Daudt LE, Fernandes JF, Vieira AK, Darrigo Junior LG, Gomes AA, Arcuri L, Lenzi L, Picharski GL, Ribeiro RC, de Figueiredo BC. Allogeneic Hematopoietic Stem Cell Transplantation for Children and Adolescents with Acute Myeloid Leukemia in Brazil: A Multicentric Retrospective Study. Cell Transplant 2020; 29:963689720949175. [PMID: 32787568 PMCID: PMC7563924 DOI: 10.1177/0963689720949175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The survival rates of children with high-risk acute myeloid leukemia (AML)
treated with hematopoietic stem cell transplant (HSCT) range from 60% to 70% in
high-income countries. The corresponding rate for Brazilian children with AML
who undergo HSCT is unknown. We conducted a retrospective analysis of 114
children with AML who underwent HSCT between 2008 and 2012 at institutions
participating in the Brazilian Pediatric Bone Marrow Transplant Working Group.
At transplant, 38% of the children were in first complete remission (CR1), 37%
were in CR2, and 25% were in CR3+ or had persistent disease. The donors included
49 matched-related, 59 matched-unrelated, and six haploidentical donors. The
most frequent source of cells was bone marrow (69%), followed by the umbilical
cord (19%) and peripheral blood (12%). The 4-year overall survival was 47% (95%
confidence interval [CI] 30%–57%), and the 4-year progression-free survival was
40% (95% CI 30%–49%). Relapse occurred in 49 patients, at a median of 122 days
after HSCT. There were 65 deaths: 40 related to AML, 19 to infection, and six to
graft versus host disease. In conclusion, our study suggests that HSCT outcomes
for children with AML in CR1 or CR2 are acceptable and that this should be
considered in the overall treatment planning for children with AML in Brazil.
Therapeutic standardization through the adoption of multicentric protocols and
appropriate supportive care treatment will have a significant impact on the
results of HSCT for AML in Brazil and possibly in other countries with limited
resources.
Collapse
Affiliation(s)
- Ana Luiza de Melo Rodrigues
- 245143Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Paraná,Brazil.,245067Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil.,176853Hospital Pequeno Príncipe, Rua Desembargador Motta, Curitiba, Paraná, Brazil
| | - Carmem Bonfim
- 176853Hospital Pequeno Príncipe, Rua Desembargador Motta, Curitiba, Paraná, Brazil.,Hospital de Clínicas da 28122Universidade Federal do Paraná, Rua General Carneiro, Curitiba, Paraná, Brazil.,417434Hospital Nossa Senhora das Graças Rua Alcídes Munhoz, Curitiba, Paraná, Brazil
| | - Adriana Seber
- 125211Hospital Samaritano, Rua Conselheiro Brotero, Higienópolis, São Paulo, Brazil
| | | | | | - Samantha Nichele
- Hospital de Clínicas da 28122Universidade Federal do Paraná, Rua General Carneiro, Curitiba, Paraná, Brazil.,417434Hospital Nossa Senhora das Graças Rua Alcídes Munhoz, Curitiba, Paraná, Brazil
| | - Liane Esteves Daudt
- 37895Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, Santa Cecilia, Porto Alegre, Rio Grande do Sul, Brazil
| | - Juliana Folloni Fernandes
- Instituto de Tratamento do Câncer Infantil Hospital de Clínicas da Faculdade de Medicina da Universidade de São Paulo, Rua Galeno de Almeida, Pinheiros, São Paulo, Brazil.,37896Hospital Israelita Albert Einstein, Jardim Leonor, São Paulo, Brazil
| | - Ana Karine Vieira
- Hospital de Clínicas da Univerdidade Federal de Minas Gerais Rua Prof. Alfredo Balena, Santa Efigenia, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Guilherme Darrigo Junior
- 54539Hospital de Clínicas da Faculdade de Medicina da Universidade de São Paulo- Ribeirão Preto, Campus Universitário, Vila Monte Alegre, Ribeirão Preto, São Paulo, Brazil
| | - Alessandra Araujo Gomes
- Instituto de Tratamento do Câncer Infantil Hospital de Clínicas da Faculdade de Medicina da Universidade de São Paulo, Rua Galeno de Almeida, Pinheiros, São Paulo, Brazil.,42522Hospital Sírio Libanês, Rua Dona Adma Jafet, Bela Vista, São Paulo, Brazil
| | - Leonardo Arcuri
- Department of Oncology and Global Pediatric Medicine, 5417St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Luana Lenzi
- Departamento de Análises Clínicas, 28122Universidade Federal do Paraná, Jardim Botanico, Curitiba, Paraná, Brazil
| | | | - Raul Correa Ribeiro
- Department of Oncology and Global Pediatric Medicine, 5417St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bonald Cavalcante de Figueiredo
- 245143Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Paraná,Brazil.,245067Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil.,Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC) at 28122Universidade Federal do Paraná, Curitiba, Paraná, Brazil.,Departamento de Saúde Coletiva, 28122Universidade Federal do Paraná, Rua Padre Camargo, Curitiba, Paraná, Brazil
| |
Collapse
|
38
|
Nucleophosmin 1 Mutations in Acute Myeloid Leukemia. Genes (Basel) 2020; 11:genes11060649. [PMID: 32545659 PMCID: PMC7348733 DOI: 10.3390/genes11060649] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022] Open
Abstract
Nucleophosmin (NPM1) is a ubiquitously expressed nucleolar protein involved in ribosome biogenesis, the maintenance of genomic integrity and the regulation of the ARF-p53 tumor-suppressor pathway among multiple other functions. Mutations in the corresponding gene cause a cytoplasmic dislocation of the NPM1 protein. These mutations are unique to acute myeloid leukemia (AML), a disease characterized by clonal expansion, impaired differentiation and the proliferation of myeloid cells in the bone marrow. Despite our improved understanding of NPM1 mutations and their consequences, the underlying leukemia pathogenesis is still unclear. Recent studies that focused on dysregulated gene expression in AML with mutated NPM1 have shed more light into these mechanisms. In this article, we review the current evidence on normal functions of NPM1 and aberrant functioning in AML, and highlight investigational strategies targeting these mutations.
Collapse
|
39
|
Dix C, Lo TH, Clark G, Abadir E. Measurable Residual Disease in Acute Myeloid Leukemia Using Flow Cytometry: A Review of Where We Are and Where We Are Going. J Clin Med 2020; 9:E1714. [PMID: 32503122 PMCID: PMC7357042 DOI: 10.3390/jcm9061714] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
The detection of measurable residual disease (MRD) has become a key investigation that plays a role in the prognostication and management of several hematologic malignancies. Acute myeloid leukemia (AML) is the most common acute leukemia in adults and the role of MRD in AML is still emerging. Prognostic markers are complex, largely based upon genetic and cytogenetic aberrations. MRD is now being incorporated into prognostic models and is a powerful predictor of relapse. While PCR-based MRD methods are sensitive and specific, many patients do not have an identifiable molecular marker. Immunophenotypic MRD methods using multiparametric flow cytometry (MFC) are widely applicable, and are based on the identification of surface marker combinations that are present on leukemic cells but not normal hematopoietic cells. Current techniques include a "different from normal" and/or a "leukemia-associated immunophenotype" approach. Limitations of MFC-based MRD analyses include the lack of standardization, the reliance on a high-quality marrow aspirate, and variable sensitivity. Emerging techniques that look to improve the detection of leukemic cells use dimensional reduction analysis, incorporating more leukemia specific markers and identifying leukemic stem cells. This review will discuss current methods together with new and emerging techniques to determine the role of MFC MRD analysis.
Collapse
Affiliation(s)
- Caroline Dix
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Tsun-Ho Lo
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW 2139, Australia; (T.-H.L.); (G.C.)
- Immunology, Sydpath, St Vincent’s Hospital, Darlinghurst, NSW 2010, Australia
| | - Georgina Clark
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW 2139, Australia; (T.-H.L.); (G.C.)
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2039, Australia
| | - Edward Abadir
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW 2139, Australia; (T.-H.L.); (G.C.)
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2039, Australia
| |
Collapse
|
40
|
Short NJ, Patel KP, Albitar M, Franquiz M, Luthra R, Kanagal-Shamanna R, Wang F, Assi R, Montalban-Bravo G, Matthews J, Ma W, Loghavi S, Takahashi K, Issa GC, Kornblau SM, Jabbour E, Garcia-Manero G, Kantarjian HM, Estrov Z, Ravandi F. Targeted next-generation sequencing of circulating cell-free DNA vs bone marrow in patients with acute myeloid leukemia. Blood Adv 2020; 4:1670-1677. [PMID: 32324887 PMCID: PMC7189293 DOI: 10.1182/bloodadvances.2019001156] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/25/2020] [Indexed: 01/19/2023] Open
Abstract
Circulating cell-free DNA (ccfDNA) allows for noninvasive peripheral blood sampling of cancer-associated mutations and has established clinical utility in several solid tumors. We performed targeted next-generation sequencing of ccfDNA and bone marrow at the time of diagnosis and after achieving remission in 22 patients with acute myeloid leukemia (AML). Among 28 genes sequenced by both platforms, a total of 39 unique somatic mutations were detected. Five mutations (13%) were detected only in ccfDNA, and 15 (38%) were detected only in bone marrow. Among the 19 mutations detected in both sources, the concordance of variant allelic frequency (VAF) assessment by both methods was high (R2 = 0.849). Mutations detected in only 1 source generally had lower VAF than those detected in both sources, suggesting that either method may miss small subclonal populations. In 3 patients, sequencing of ccfDNA detected new or persistent leukemia-associated mutations during remission that appeared to herald overt relapse. Overall, this study demonstrates that sequencing of ccfDNA in patients with AML can identify clinically relevant mutations not detected in the bone marrow and may play a role in the assessment of measurable residual disease. However, mutations were missed by both ccfDNA and bone marrow analyses, particularly when the VAF was <10%, suggesting that ccfDNA and bone marrow may be complementary in the assessment and monitoring of patients with AML.
Collapse
Affiliation(s)
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Feng Wang
- Department of Bioinformatics, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Rita Assi
- Lebanese American University Medical Center-Rizk Hospital, Beirut, Lebanon
| | | | | | | | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Short NJ, Konopleva M, Kadia TM, Borthakur G, Ravandi F, DiNardo CD, Daver N. Advances in the Treatment of Acute Myeloid Leukemia: New Drugs and New Challenges. Cancer Discov 2020; 10:506-525. [DOI: 10.1158/2159-8290.cd-19-1011] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/23/2019] [Accepted: 11/20/2019] [Indexed: 11/16/2022]
|
42
|
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-SCT) is the most established and commonly used cellular immunotherapy in cancer care. It is the most potent anti-leukemic therapy in patients with acute myeloid leukemia (AML) and is routinely used with curative intent in patients with intermediate and poor risk disease. Donor T cells, and possibly other immune cells, eliminate residual leukemia cells after prior (radio)chemotherapy. This immune-mediated response is known as graft-versus-leukemia (GvL). Donor alloimmune responses can also be directed against healthy tissues, which is known as graft-versus-host disease (GvHD). GvHD and GvL often co-occur and, therefore, a major barrier to exploiting the full immunotherapeutic benefit of donor immune cells against patient leukemia is the immunosuppression required to treat GvHD. However, curative responses to allo-SCT and GvHD do not always occur together, suggesting that these two immune responses could be de-coupled in some patients. To make further progress in successfully promoting GvL without GvHD, we must transform our limited understanding of the cellular and molecular basis of GvL and GvHD. Specifically, in most patients we do not understand the antigenic basis of immune responses in GvL and GvHD. Identification of antigens important for GvL but not GvHD, and vice versa, could impact on donor selection, allow us to track GvL immune responses and begin to specifically harness and strengthen anti-leukemic immune responses against patient AML cells, whilst minimizing the toxicity of GvHD.
Collapse
Affiliation(s)
- Connor Sweeney
- MRC Molecular Haematology Unit, Oxford Biomedical Research Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Paresh Vyas
- MRC Molecular Haematology Unit, Oxford Biomedical Research Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|