1
|
Shimony S, Stahl M, Stone RM. Acute Myeloid Leukemia: 2025 Update on Diagnosis, Risk-Stratification, and Management. Am J Hematol 2025; 100:860-891. [PMID: 39936576 PMCID: PMC11966364 DOI: 10.1002/ajh.27625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/13/2025]
Abstract
DISEASE OVERVIEW Acute myeloid leukemia (AML) is a bone marrow stem cell cancer that is often fatal despite available treatments. Diagnosis, risk assessment, monitoring, and therapeutic management of AML have changed dramatically in the last decade due to increased pathophysiologic understanding, improved assessment technology, and the addition of at least 12 approved therapies. DIAGNOSIS The diagnosis is based on the presence of immature leukemia cells in the blood, and/or bone marrow or less often in extra-medullary tissues. New biological insights have been integrated into recent classification systems. RISK ASSESSMENT The European Leukemia Network has published risk classification algorithms for both intensively and non-intensively treated patients based on cytogenetic and on molecular findings. Prognostic factors may differ based on the therapeutic approach. MONITORING Our increasing ability to quantify lower levels of measurable residual disease (MRD) potentially allows better response assessment, as well as dynamic monitoring of disease status. The incorporation of MRD findings into therapeutic decision-making is rapidly evolving. RISK ADAPTED THERAPY The availability of 12 newly approved agents has been welcomed; however, optimal strategies incorporating newer agents into therapeutic algorithms are debated. The overarching approach integrates patient and caregiver goals of care, comorbidities, and disease characteristics.
Collapse
Affiliation(s)
- Shai Shimony
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Maximilian Stahl
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Richard M. Stone
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| |
Collapse
|
2
|
Gao Y, Li J, Wang N, An W, Yin Z, Wang J, Chen X, Chen Y, Guo Y, Yang W, Zhang L, Zou Y, Chen X, Zhu X. TP53 deletion as an MRD-dependent risk factor in childhood B-ALL: A post hoc analysis from a prospective cohort. Hemasphere 2025; 9:e70115. [PMID: 40171518 PMCID: PMC11956791 DOI: 10.1002/hem3.70115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 04/03/2025] Open
Abstract
The effect of TP53 alterations on childhood B-cell acute lymphoblastic leukemia (B-ALL) remains unclear. To investigate the prognostic value of TP53 deletion (TP53 del ) and TP53 mutation (TP53 mut ), this post hoc study used fluorescence in situ hybridization test to detect TP53 del in 907 newly diagnosed B-ALL patients from a prospective cohort of Chinese Children's Cancer Group ALL-2015 trial. Targeted gene sequencing was used to identify TP53 mut in 342 out of the 907 patients. TP53 del was detected in 4.4% of patients. The frequency of hypodiploidy was higher in TP53 del subgroup (7.5% vs. 0.5%, p = 0.002), but patients with TP53 del were less likely to have other recurrent genetic abnormalities, including BCR::ABL1, ETV6::RUNX1, TCF3::PBX1 and KMT2A rearrangements. Univariable and multivariable analyses indicated that TP53 del was an independent risk factor for overall survival (OS) and disease-free survival (DFS). Furthermore, stratification analysis revealed that TP53 del was associated with lower 5-year DFS in patients with positive minimal residual disease (MRD) after induction in the intermediate-risk group (0.0% vs. 58.0% [95% confidence interval [CI] 49.2%-68.3%], p < 0.001), suggesting an MRD-dependent pattern. However, somatic TP53 mut was not associated with poor survival (81.8% [95% CI 61.9%-100.0%] vs. 84.9% [95% CI 81.1%-89.0%], p = 0.971). In summary, TP53 del may serve as a predictor for poor prognosis in pediatric B-ALL. In particular, children in the intermediate-risk group with positive MRD and TP53 del may require more aggressive treatment.
Collapse
Affiliation(s)
- Yangyang Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Jun Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Ning Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Wenbin An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Zixi Yin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Junxia Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Xia Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Yumei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Ye Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Wenyu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Li Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Yao Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Xiaojuan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Tianjin Institutes of Health ScienceTianjinChina
| |
Collapse
|
3
|
Fikar P, Alvarez L, Berne L, Cienciala M, Kan C, Kasl H, Luo M, Novackova Z, Ordonez S, Sramkova Z, Holubova M, Lysak D, Avery L, Caro AA, Crowder RN, Diaz-Martinez LA, Donley DW, Giorno RR, Reed IKG, Hensley LL, Johnson KC, Kim AY, Kim P, LaGier AJ, Newman JJ, Padilla-Crespo E, Reyna NS, Tsotakos N, Al-Saadi NN, Appleton T, Arosemena-Pickett A, Bell BA, Bing G, Bishop B, Forde C, Foster MJ, Gray K, Hasley BL, Johnson K, Jones DJ, LaShall AC, McGuire K, McNaughton N, Morgan AM, Norris L, Ossman LA, Rivera-Torres PA, Robison ME, Thibodaux K, Valmond L, Georgiev D. Enhancing reproducibility in single cell research with biocytometry: An inter-laboratory study. PLoS One 2024; 19:e0314992. [PMID: 39652549 PMCID: PMC11627387 DOI: 10.1371/journal.pone.0314992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Biomedicine today is experiencing a shift towards decentralized data collection, which promises enhanced reproducibility and collaboration across diverse laboratory environments. This inter-laboratory study evaluates the performance of biocytometry, a method utilizing engineered bioparticles for enumerating cells based on their surface antigen patterns. In centralized and aggregated inter-lab studies, biocytometry demonstrated significant statistical power in discriminating numbers of target cells at varying concentrations as low as 1 cell per 100,000 background cells. User skill levels varied from expert to beginner capturing a range of proficiencies. Measurement was performed in a decentralized environment without any instrument cross-calibration or advanced user training outside of a basic instruction manual. The results affirm biocytometry to be a viable solution for immunophenotyping applications demanding sensitivity as well as scalability and reproducibility and paves the way for decentralized analysis of rare cells in heterogeneous samples.
Collapse
Affiliation(s)
- Pavel Fikar
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Laura Alvarez
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Laura Berne
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Martin Cienciala
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Christopher Kan
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Hynek Kasl
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Mona Luo
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Zuzana Novackova
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Sheyla Ordonez
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Zuzana Sramkova
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
| | - Monika Holubova
- Department of Hematology and Oncology, University Hospital Pilsen, Pilsen, Czech Republic
| | - Daniel Lysak
- Department of Hematology and Oncology, University Hospital Pilsen, Pilsen, Czech Republic
| | - Lyndsay Avery
- Department of Biology, Saint Michael’s College, Colchester, Vermont, United States of America
| | - Andres A. Caro
- Chemistry Department, Hendrix College, Conway, Arkansas, United States of America
| | - Roslyn N. Crowder
- Department of Biology, Stetson University, Deland, Florida, United States of America
| | - Laura A. Diaz-Martinez
- Department of Biology, Gonzaga University, Spokane, Washington, United States of America
| | - David W. Donley
- Department of Biology, Harding University, Searcy, Arkansas, United States of America
| | - Rebecca R. Giorno
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Irene K. Guttilla Reed
- Department of Biology, University of Saint Joseph, West Hartford, Connecticut, United States of America
| | - Lori L. Hensley
- Department of Biological Sciences, Jacksonville State University, Jacksonville, Alabama, United States of America
| | - Kristen C. Johnson
- Department of Life Sciences, University of New Hampshire, Manchester, New Hampshire, United States of America
| | - Audrey Y. Kim
- Department of Biological Sciences, Grambling State University, Grambling, Louisiana, United States of America
| | - Paul Kim
- Department of Biological Sciences, Grambling State University, Grambling, Louisiana, United States of America
| | - Adriana J. LaGier
- Department of Biology, College of Social and Natural Sciences, Grand View University, Des Moines, Iowa, United States of America
| | - Jamie J. Newman
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Elizabeth Padilla-Crespo
- Department of Science and Technology, Inter American University of Puerto Rico-Aguadilla, Aguadilla, Puerto Rico, United States of America
| | - Nathan S. Reyna
- Department of Biology, Ouachita Baptist University, Arkadelphia, Arkansas, United States of America
| | - Nikolaos Tsotakos
- Department of Biological Sciences, School of Science Engineering and Technology, Penn State Harrisburg, Middletown, Pennsylvania, United States of America
| | - Noha N. Al-Saadi
- Department of Biological Sciences, Jacksonville State University, Jacksonville, Alabama, United States of America
| | - Tayler Appleton
- Department of Biology, Harding University, Searcy, Arkansas, United States of America
| | - Ana Arosemena-Pickett
- Department of Science and Technology, Inter American University of Puerto Rico-Aguadilla, Aguadilla, Puerto Rico, United States of America
| | - Braden A. Bell
- Department of Biology, Gonzaga University, Spokane, Washington, United States of America
| | - Grace Bing
- Department of Biology, Harding University, Searcy, Arkansas, United States of America
| | - Bre Bishop
- Department of Biology, Harding University, Searcy, Arkansas, United States of America
| | - Christa Forde
- Department of Biological Sciences, Grambling State University, Grambling, Louisiana, United States of America
| | - Michael J. Foster
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Kassidy Gray
- Department of Biology, Ouachita Baptist University, Arkadelphia, Arkansas, United States of America
| | - Bennett L. Hasley
- Department of Biological Sciences, Jacksonville State University, Jacksonville, Alabama, United States of America
| | - Kennedy Johnson
- Department of Biology, Ouachita Baptist University, Arkadelphia, Arkansas, United States of America
| | - Destiny J. Jones
- Department of Biological Sciences, Grambling State University, Grambling, Louisiana, United States of America
| | - Allison C. LaShall
- Department of Biological Sciences, Jacksonville State University, Jacksonville, Alabama, United States of America
| | - Kennedy McGuire
- Department of Biology, Ouachita Baptist University, Arkadelphia, Arkansas, United States of America
| | - Naomi McNaughton
- Department of Biology, Harding University, Searcy, Arkansas, United States of America
| | - Angelina M. Morgan
- Department of Biological Sciences, Grambling State University, Grambling, Louisiana, United States of America
| | - Lucas Norris
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Landon A. Ossman
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Paollette A. Rivera-Torres
- Department of Science and Technology, Inter American University of Puerto Rico-Aguadilla, Aguadilla, Puerto Rico, United States of America
| | - Madeline E. Robison
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Kathryn Thibodaux
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Lescia Valmond
- Department of Biological Sciences, Grambling State University, Grambling, Louisiana, United States of America
| | - Daniel Georgiev
- Department of Research & Development, Sampling Human Inc., Berkeley, California, United States of America
- Faculty of Applied Sciences, University of West Bohemia, Pilsen, Czech Republic
| |
Collapse
|
4
|
Álvarez Flores MB, Sopeña Corvinos M, Guillén Santos R, Cava Valenciano F. Fluorescent Aerolysin (FLAER) Binding Is Abnormally Low in the Clonal Precursors of Acute Leukemias, with Binding Particularly Low or Absent in Acute Promyelocytic Leukemia. Int J Mol Sci 2024; 25:11898. [PMID: 39595968 PMCID: PMC11593337 DOI: 10.3390/ijms252211898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Flow cytometry plays a fundamental role in the diagnosis of leukemias and lymphomas, as well as in the follow-up and evaluation of minimally measurable disease after treatment. In some instances, such as in the case of acute promyelocytic leukemia (APL), rapid diagnosis is required to avoid death due to serious blood clotting or bleeding complications. Given that promyelocytes do not express the glycophosphatidylinositol (GPI)-anchored protein CD16 and that deficient CD16 expression is a feature of some CD16 polymorphisms and paroxysmal nocturnal hemoglobinuria (PNH), we included the GPI anchor probe FLAER aerolysin in the APL flow cytometry probe panel. Initial tests showed that FLAER binding was absent in pathological promyelocytes from APL patients but was consistently detected with high intensity in healthy promyelocytes from control bone marrow. FLAER binding was studied in 71 hematologic malignancies. Appropriate control cells were obtained from 16 bone marrow samples from patients with idiopathic thrombocytopenic purpura and non-infiltrated non-Hodgkin's lymphoma. Compared with the positive FLAER signal in promyelocytes from healthy bone marrow, malignant promyelocytes from APL patients showed weak or negative FLAER binding. The FLAER signal in APL promyelocytes was also lower than that in control myeloid progenitors and precursors from patients with other forms of acute myeloid leukemia (AML), B-cell acute lymphoblastic leukemia, or myelodysplastic syndrome. Minimal measurable disease studies performed in APL patients after treatment found normal promyelocyte expression when minimal measurable disease was negative and FLAER-negative promyelocytes when disease relapse was detected. The inclusion of FLAER in the flow cytometry diagnosis and follow-up of APL could be very helpful. Decreased FLAER binding was found in all cases of APL, confirmed by the detection of the PML-RARA fusion transcript and, to a lesser extent, in the other AMLs studied. This study also revealed FLAER differences in other acute leukemias and even between different precursors (myeloid and lymphoid) from healthy controls. However, the reason for FLAER's non-binding to the malignant precursors of these leukemias remains unknown, and future studies should explore the possible relation with an immune escape phenomenon in these leukemias.
Collapse
MESH Headings
- Humans
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/diagnosis
- Leukemia, Promyelocytic, Acute/pathology
- Pore Forming Cytotoxic Proteins/metabolism
- Pore Forming Cytotoxic Proteins/genetics
- Bacterial Toxins/metabolism
- Bacterial Toxins/genetics
- Flow Cytometry
- Male
- Female
- Adult
- Middle Aged
- Aged
- Receptors, IgG/metabolism
- Receptors, IgG/genetics
- Protein Binding
Collapse
Affiliation(s)
| | - María Sopeña Corvinos
- URSalud Laboratory, Hospital Universitario Infanta Sofia, 28702 San Sebastián de los Reyes, Spain; (M.S.C.); (R.G.S.); (F.C.V.)
| | - Raquel Guillén Santos
- URSalud Laboratory, Hospital Universitario Infanta Sofia, 28702 San Sebastián de los Reyes, Spain; (M.S.C.); (R.G.S.); (F.C.V.)
| | - Fernando Cava Valenciano
- URSalud Laboratory, Hospital Universitario Infanta Sofia, 28702 San Sebastián de los Reyes, Spain; (M.S.C.); (R.G.S.); (F.C.V.)
| |
Collapse
|
5
|
Hartzell CM, Shaver AC, Mason EF. Flow Cytometric Assessment of Malignant Hematologic Disorders. Clin Lab Med 2024; 44:465-477. [PMID: 39089752 DOI: 10.1016/j.cll.2024.04.008] [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] [Indexed: 08/04/2024]
Abstract
Multiparameter flow cytometry (MPF) is an essential component of the diagnostic workup of hematologic malignancies. Recently developed tools have expanded the utility of MPF in detecting T-cell clonality and myelomonocytic dysplasia. Minimal/measurable residual disease analysis has long been established as critical in the management of B-lymphoblastic leukemia and is emerging as a useful tool in myeloid malignancies. With the continued increased complexity of MPF assays, emerging tools for data collection and analysis will allow users to take full advantage of MPF in the diagnosis of hematologic disease.
Collapse
Affiliation(s)
- Connor M Hartzell
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, 445 Great Circle Road, Nashville, TN 37228, USA
| | - Aaron C Shaver
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, 445 Great Circle Road, Nashville, TN 37228, USA
| | - Emily F Mason
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, 445 Great Circle Road, Nashville, TN 37228, USA.
| |
Collapse
|
6
|
Tran V, Salafian K, Michaels K, Jones C, Reed D, Keng M, El Chaer F. MRD in Philadelphia Chromosome-Positive ALL: Methodologies and Clinical Implications. Curr Hematol Malig Rep 2024; 19:186-196. [PMID: 38888822 DOI: 10.1007/s11899-024-00736-9] [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] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE OF REVIEW Measurable residual disease (MRD) is integral in the management of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). This review discusses the current methods used to evaluate MRD as well as the interpretation, significance, and incorporation of MRD in current practice. RECENT FINDINGS New molecular technologies have allowed the detection of MRD to levels as low as 10- 6. The most used techniques to evaluate MRD are multiparametric flow cytometry (MFC), quantitative reverse transcription polymerase chain reaction (RT-qPCR), and high-throughput next-generation sequencing (NGS). Each method varies in terms of advantages, disadvantages, and MRD sensitivity. MRD negativity after induction treatment and after allogeneic hematopoietic cell transplantation (HCT) is an important prognostic marker that has consistently been shown to be associated with improved outcomes. Blinatumomab, a new targeted therapy for Ph + ALL, demonstrates high efficacy in eradicating MRD and improving patient outcomes. In the relapsed/refractory setting, the use of inotuzumab ozogamicin and tisagenlecleucel has shown promise in eradicating MRD. The presence of MRD has become an important predictive measure in Ph + ALL. Current studies evaluate the use of MRD in treatment decisions, especially in expanding therapeutic options for Ph + ALL, including tyrosine kinase inhibitors, targeted antibody therapies, chimeric antigen receptor cell therapy, and HCT.
Collapse
Affiliation(s)
- Valerie Tran
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Kiarash Salafian
- Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Kenan Michaels
- Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Caroline Jones
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Daniel Reed
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Michael Keng
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Firas El Chaer
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
7
|
Lebecque B, Besombes J, Dannus LT, De Antonio M, Cacheux V, Grèze V, Montagnon V, Veronese L, Tchirkov A, Tournilhac O, Berger MG, Veyrat-Masson R. Faster clinical decisions in B-cell acute lymphoblastic leukaemia: A single flow cytometric 12-colour tube improves diagnosis and minimal residual disease follow-up. Br J Haematol 2024; 204:1872-1881. [PMID: 38432068 DOI: 10.1111/bjh.19390] [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: 01/12/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Assessing minimal residual disease (MRD) in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) is essential for adjusting therapeutic strategies and predicting relapse. Quantitative polymerase chain reaction (qPCR) is the gold standard for MRD. Alternatively, flow cytometry is a quicker and cost-effective method that typically uses leukaemia-associated immunophenotype (LAIP) or different-from-normal (DFN) approaches for MRD assessment. This study describes an optimized 12-colour flow cytometry antibody panel designed for BCP-ALL diagnosis and MRD monitoring in a single tube. This method robustly differentiated hematogones and BCP-ALL cells using two specific markers: CD43 and CD81. These and other markers (e.g. CD73, CD66c and CD49f) enhanced the specificity of BCP-ALL cell detection. This innovative approach, based on a dual DFN/LAIP strategy with a principal component analysis method, can be used for all patients and enables MRD analysis even in the absence of a diagnostic sample. The robustness of our method for MRD monitoring was confirmed by the strong correlation (r = 0.87) with the qPCR results. Moreover, it simplifies and accelerates the preanalytical process through the use of a stain/lysis/wash method within a single tube (<2 h). Our flow cytometry-based methodology improves the BCP-ALL diagnosis efficiency and MRD management, offering a complementary method with considerable benefits for clinical laboratories.
Collapse
Affiliation(s)
- Benjamin Lebecque
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Joevin Besombes
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Louis-Thomas Dannus
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Marie De Antonio
- Unité de Biostatistiques, Direction de la Recherche Clinique et de l'Innovation, Centre Hospitalier Universitaire de Clermont-Ferrand, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Victoria Cacheux
- Service de Thérapie Cellulaire et Hématologie Clinique Adulte, Clermont-Ferrand, France
| | - Victoria Grèze
- CHU Clermont-Ferrand, Service Hématologie Oncologie Pédiatrique, Hôpital Estaing, Clermont-Ferrand, France
| | - Valentin Montagnon
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
| | - Lauren Veronese
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, Clermont-Ferrand, France
| | - Andrei Tchirkov
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, Clermont-Ferrand, France
| | - Olivier Tournilhac
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
- Service de Thérapie Cellulaire et Hématologie Clinique Adulte, Clermont-Ferrand, France
| | - Marc G Berger
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
| | | |
Collapse
|
8
|
Tettero JM, Heidinga ME, Mocking TR, Fransen G, Kelder A, Scholten WJ, Snel AN, Ngai LL, Bachas C, van de Loosdrecht AA, Ossenkoppele GJ, de Leeuw DC, Cloos J, Janssen JJWM. Impact of hemodilution on flow cytometry based measurable residual disease assessment in acute myeloid leukemia. Leukemia 2024; 38:630-639. [PMID: 38272991 PMCID: PMC10912027 DOI: 10.1038/s41375-024-02158-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/27/2024]
Abstract
Measurable residual disease (MRD) measured in the bone marrow (BM) of acute myeloid leukemia (AML) patients after induction chemotherapy is an established prognostic factor. Hemodilution, stemming from peripheral blood (PB) mixing within BM during aspiration, can yield false-negative MRD results. We prospectively examined hemodilution by measuring MRD in BM aspirates obtained from three consecutive 2 mL pulls, along with PB samples. Our results demonstrated a significant decrease in MRD percentages between the first and second pulls (P = 0.025) and between the second and third pulls (P = 0.025), highlighting the impact of hemodilution. Initially, 39% of MRD levels (18/46 leukemia-associated immunophenotypes) exceeded the 0.1% cut-off, decreasing to 30% (14/46) in the third pull. Additionally, we assessed the performance of six published methods and parameters for distinguishing BM from PB samples, addressing or compensating for hemodilution. The most promising results relied on the percentages of CD16dim granulocytic population (scarce in BM) and CD117high mast cells (exclusive to BM). Our findings highlight the importance of estimating hemodilution in MRD assessment to qualify MRD results, particularly near the common 0.1% cut-off. To avoid false-negative results by hemodilution, it is essential to collect high-quality BM aspirations and preferably utilizing the initial pull for MRD testing.
Collapse
Affiliation(s)
- Jesse M Tettero
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Maaike E Heidinga
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Tim R Mocking
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Glenn Fransen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Angèle Kelder
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Willemijn J Scholten
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Alexander N Snel
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Lok Lam Ngai
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Costa Bachas
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Arjan A van de Loosdrecht
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Gert J Ossenkoppele
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - David C de Leeuw
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| | - Jeroen J W M Janssen
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Lin M, Zhao X, Chang Y, Zhao X. Current assessment and management of measurable residual disease in patients with acute lymphoblastic leukemia in the setting of CAR-T-cell therapy. Chin Med J (Engl) 2024; 137:140-151. [PMID: 38148315 PMCID: PMC10798764 DOI: 10.1097/cm9.0000000000002945] [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: 06/18/2023] [Indexed: 12/28/2023] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR)-modified T-cell therapy has achieved remarkable success in the treatment of acute lymphoblastic leukemia (ALL). Measurable/minimal residual disease (MRD) monitoring plays a significant role in the prognostication and management of patients undergoing CAR-T-cell therapy. Common MRD detection methods include flow cytometry (FCM), polymerase chain reaction (PCR), and next-generation sequencing (NGS), and each method has advantages and limitations. It has been well documented that MRD positivity predicts a poor prognosis and even disease relapse. Thus, how to perform prognostic evaluations, stratify risk based on MRD status, and apply MRD monitoring to guide individual therapeutic decisions have important implications in clinical practice. This review assesses the common and novel MRD assessment methods. In addition, we emphasize the critical role of MRD as a prognostic biomarker and summarize the latest studies regarding MRD-directed combination therapy with CAR-T-cell therapy and allogeneic hematopoietic stem cell transplantation (allo-HSCT), as well as other therapeutic strategies to improve treatment effect. Furthermore, this review discusses current challenges and strategies for MRD detection in the setting of disease relapse after targeted therapy.
Collapse
Affiliation(s)
- Minghao Lin
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiaosu Zhao
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yingjun Chang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiangyu Zhao
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| |
Collapse
|
10
|
Chatterjee G, Dhende P, Raj S, Shetty V, Ghogale S, Deshpande N, Girase K, Patil J, Kalra A, Narula G, Dalvi K, Dhamne C, Moulik NR, Rajpal S, Patkar NV, Banavali S, Gujral S, Subramanian PG, Tembhare PR. 15-color highly sensitive flow cytometry assay for post anti-CD19 targeted therapy (anti-CD19-CAR-T and blinatumomab) measurable residual disease assessment in B-lymphoblastic leukemia/lymphoma: Real-world applicability and challenges. Eur J Haematol 2024; 112:122-136. [PMID: 37706583 DOI: 10.1111/ejh.14102] [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: 06/21/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
OBJECTIVES Measurable residual disease (MRD) is the most relevant predictor of disease-free survival in B-cell acute lymphoblastic leukemia (B-ALL). We aimed to establish a highly sensitive flow cytometry (MFC)-based B-ALL-MRD (BMRD) assay for patients receiving anti-CD19 immunotherapy with an alternate gating approach and to document the prevalence and immunophenotype of recurrently occurring low-level mimics and confounding populations. METHODS We standardized a 15-color highly-sensitive BMRD assay with an alternate CD19-free gating approach. The study included 137 MRD samples from 43 relapsed/refractory B-ALL patients considered for anti-CD19 immunotherapy. RESULTS The 15-color BMRD assay with CD22/CD24/CD81/CD33-based gating approach was routinely applicable in 137 BM samples and could achieve a sensitivity of 0.0005%. MRD was detected in 29.9% (41/137) samples with 31.7% (13/41) of them showing <.01% MRD. Recurrently occurring low-level cells that showed immunophenotypic overlap with leukemic B-blasts included: (a) CD19+CD10+CD34+CD22+CD24+CD81+CD123+CD304+ plasmacytoid dendritic cells, (b) CD73bright/CD304bright/CD81bright mesenchymal stromal/stem cells (CD10+) and endothelial cells (CD34+CD24+), (c) CD22dim/CD34+/CD38dim/CD81dim/CD19-/CD10-/CD24- early lymphoid progenitor/precursor type-1 cells (ELP-1) and (d) CD22+/CD34+/CD10heterogeneous/CD38moderate/CD81moderate/CD19-/CD24- stage-0 B-cell precursors or ELP-2 cells. CONCLUSIONS We standardized a highly sensitive 15-color BMRD assay with a non-CD19-based gating strategy for patients receiving anti-CD19 immunotherapy. We also described the immunophenotypes of recurrently occurring low-level populations that can be misinterpreted as MRD in real-world practice.
Collapse
Affiliation(s)
- Gaurav Chatterjee
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Priyanka Dhende
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Simpy Raj
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Vruksha Shetty
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Sitaram Ghogale
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Nilesh Deshpande
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Karishma Girase
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Jagruti Patil
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Aastha Kalra
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Gaurav Narula
- Department of Pediatric Oncology, Tata Memorial Center, Mumbai, Mumbai, Maharashtra, India
| | - Kajal Dalvi
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Chetan Dhamne
- Department of Pediatric Oncology, Tata Memorial Center, Mumbai, Mumbai, Maharashtra, India
| | - Nirmalya Roy Moulik
- Department of Pediatric Oncology, Tata Memorial Center, Mumbai, Mumbai, Maharashtra, India
| | - Sweta Rajpal
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Nikhil V Patkar
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Shripad Banavali
- Department of Pediatric Oncology, Tata Memorial Center, Mumbai, Mumbai, Maharashtra, India
| | - Sumeet Gujral
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Mumbai, Maharashtra, India
| | - Papagudi G Subramanian
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Prashant R Tembhare
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| |
Collapse
|
11
|
Li S, Wang X, Liu L, Liu J, Rao J, Yuan Z, Gao L, Li Y, Luo L, Li G, Li Z, Li S, He J, Zhang L, Chen L, Huang W, Yin P, Li C, Li X, Wang Y, Dong Y, Zhang D, Zang Q, Chen Y, Shen L, Li W, Cao W, Zhang X, Wang S. CD7 targeted "off-the-shelf" CAR-T demonstrates robust in vivo expansion and high efficacy in the treatment of patients with relapsed and refractory T cell malignancies. Leukemia 2023; 37:2176-2186. [PMID: 37700087 DOI: 10.1038/s41375-023-02018-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 08/17/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) represents an area of highly unmet medical needs. Once relapsed, patients have limited treatment options and poor prognosis. T-ALL antigens such as CD7 is extensively expressed in normal T cells and natural killer (NK) cells, and extending the success of CAR-T therapy to T cell malignancies was challenged by CAR-T cell fratricide, high production cost, and potential product contaminations. GC027 is an "off-the-shelf" allogeneic CD7 targeted CAR-T therapeutic product for T cell malignancies. It demonstrated superior cell expansion and antileukemia efficacy in mouse xenograft model. In our previous study, we observed promising efficacy results in the first two relapsed and refractory(R/R) T-ALL patients treated with GC027. In the expanded study, 11 out of 12 patients had rapid eradication of T-lymphoblasts and reached complete response within 1-month after GC027 infusion. GC027 cells expanded quickly beginning at infusion and reached to peak around 5-10 days after infusion. For most patients with a response(9/11), GC027 could not be detected via flow cytometry or qPCR 4 weeks after infusion. One patient had progression free survival of >3 years. With manageable toxicity profile, GC027 demonstrated superior clinical efficacy to standard chemotherapy regimens in (R/R) T cell malignancies.
Collapse
Affiliation(s)
- Shiqi Li
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Xinxin Wang
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Lin Liu
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Jia Liu
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Jun Rao
- Xinqiao Hospital, Chongqing, 400037, China
| | - Zhongtao Yuan
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Li Gao
- Xinqiao Hospital, Chongqing, 400037, China
| | - Yu Li
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Le Luo
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Gui Li
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Zhimin Li
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Sheng Li
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Jiaping He
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Lihua Zhang
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Lvzhe Chen
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Wenhui Huang
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Ping Yin
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Chunmin Li
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Xiaoping Li
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Youcheng Wang
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Yancheng Dong
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Dingsong Zhang
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Qingying Zang
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Yingnian Chen
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China
| | - Lianjun Shen
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Wenling Li
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China
| | - Wei Cao
- Gracell Biotechnologies Co., Ltd, Shanghai, 200030, China.
| | - Xi Zhang
- Xinqiao Hospital, Chongqing, 400037, China.
| | - Sanbin Wang
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, 650100, Yunnan, China.
| |
Collapse
|
12
|
Kanaan SB, Urselli F, Radich JP, Nelson JL. Ultrasensitive chimerism enhances measurable residual disease testing after allogeneic hematopoietic cell transplantation. Blood Adv 2023; 7:6066-6079. [PMID: 37467017 PMCID: PMC10582300 DOI: 10.1182/bloodadvances.2023010332] [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: 04/04/2023] [Revised: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023] Open
Abstract
Increasing mixed chimerism (reemerging recipient cells) after allogeneic hematopoietic cell transplant (allo-HCT) can indicate relapse, the leading factor determining mortality in blood malignancies. Most clinical chimerism tests have limited sensitivity and are primarily designed to monitor engraftment. We developed a panel of quantitative polymerase chain reaction assays using TaqMan chemistry capable of quantifying chimerism in the order of 1 in a million. At such analytic sensitivity, we hypothesized that it could inform on relapse risk. As a proof-of-concept, we applied our panel to a retrospective cohort of patients with acute leukemia who underwent allo-HCT with known outcomes. Recipient cells in bone marrow aspirates (BMAs) remained detectable in 97.8% of tested samples. Absolute recipient chimerism proportions and rates at which these proportions increased in BMAs in the first 540 days after allo-HCT were associated with relapse. Detectable measurable residual disease (MRD) via flow cytometry in BMAs after allo-HCT showed limited correlation with relapse. This correlation noticeably strengthened when combined with increased recipient chimerism in BMAs, demonstrating the ability of our ultrasensitive chimerism assay to augment MRD data. Our technology reveals an underappreciated usefulness of clinical chimerism. Used side by side with MRD assays, it promises to improve identification of patients with the highest risk of disease reoccurrence for a chance of early intervention.
Collapse
Affiliation(s)
- Sami B. Kanaan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Research and Development, Chimerocyte Inc, Seattle, WA
| | - Francesca Urselli
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jerald P. Radich
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - J. Lee Nelson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Research and Development, Chimerocyte Inc, Seattle, WA
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
13
|
van der Linde R, Gatt PN, Smith S, Fernandez MA, Vaughan L, Blyth E, Curnow J, Brown DA, Tegg E, Sasson SC. Measurable Residual Disease (MRD) by Flow Cytometry in Adult B-Acute Lymphoblastic Leukaemia (B-ALL) and Acute Myeloid Leukaemia (AML): Correlation with Molecular MRD Testing and Clinical Outcome at One Year. Cancers (Basel) 2023; 15:5064. [PMID: 37894431 PMCID: PMC10605425 DOI: 10.3390/cancers15205064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Measurable residual disease (MRD) detected by flow cytometry (FC) is well established in paediatric B- lymphoblastic leukaemia (B-ALL) and adult chronic lymphocytic leukaemia (CLL), but its utility in adult B-ALL and adult acute myeloid leukaemia (AML) is less clear. In this prospective MRD study, one of the largest in Australia to date, we examined consecutive bone marrow aspirates from adult participants with B-ALL (n = 47) and AML (n = 87) sent for FC-MRD testing at a quaternary referral hospital in Sydney. FC-MRD results were correlated to corresponding Mol-MRD testing where available and clinical outcomes at three-month intervals over 1 year. B-ALL showed a moderate positive correlation (rs = 0.401, p < 0.001), while there was no correlation between FC-MRD and Mol-MRD for AML (rs = 0.13, p = 0.237). Five FC-MRD patterns were identified which had significant associations with relapse (X2(4) = 31.17(4), p > 0.001) and survival (X2(4) = 13.67, p = 0.008) in AML, but not in B-ALL. The three-month MRD results were also strongly associated with survival in AML, while the association in B-ALL was less evident. There was a moderate correlation between FC-MRD and Mol-MRD in B-ALL but not AML. The association of FC-MRD with relapse and survival was stronger in AML than in B-ALL. Overall, these findings suggest divergent utilities of FC-MRD in AML and B-ALL.
Collapse
Affiliation(s)
- Riana van der Linde
- Department of Laboratory Haematology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (L.V.); (E.T.)
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
| | - Prudence N. Gatt
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Westmead Institute for Medical Research, University of Sydney, Sydney, NSW 2145, Australia
| | - Sandy Smith
- Flow Cytometry Unit, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (S.S.); (M.A.F.)
| | - Marian A. Fernandez
- Flow Cytometry Unit, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (S.S.); (M.A.F.)
| | - Lachlin Vaughan
- Department of Laboratory Haematology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (L.V.); (E.T.)
- Department of Haematology, Western Sydney Local Health District, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Emily Blyth
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Westmead Institute for Medical Research, University of Sydney, Sydney, NSW 2145, Australia
- Department of Haematology, Western Sydney Local Health District, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Jennifer Curnow
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Department of Haematology, Western Sydney Local Health District, Westmead Hospital, Westmead, NSW 2145, Australia
| | - David A. Brown
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Westmead Institute for Medical Research, University of Sydney, Sydney, NSW 2145, Australia
- Department of Clinical Immunology and Immunopathology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Elizabeth Tegg
- Department of Laboratory Haematology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (L.V.); (E.T.)
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
| | - Sarah C. Sasson
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Department of Clinical Immunology and Immunopathology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia
| |
Collapse
|
14
|
Liao H, Jiang N, Yang Y, Zhang X, Chen J, Lai H, Zheng Q. Association of Minimal Residual Disease by a Single-Tube 8-Color Flow Cytometric Analysis With Clinical Outcome in Adult B-Cell Acute Lymphoblastic Leukemia: A Real-World Study Based on 486 Patients. Arch Pathol Lab Med 2023; 147:1186-1195. [PMID: 36508349 DOI: 10.5858/arpa.2022-0172-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2022] [Indexed: 09/29/2023]
Abstract
CONTEXT.— Minimal/measurable residual disease (MRD) measured by molecular and multiparametric flow cytometry (MFC) has been proven to be predictive of relapse and survival in patients with B-cell acute lymphoblastic leukemia (B-ALL). A universally applicable antibody panel at a low cost but without compromising sensitivity and power of prognosis prediction in adult B-ALL remains unestablished. OBJECTIVE.— To report our experience of using a single-tube 8-color MFC panel to measure the MRD status as a prognostic indicator in adult B-ALL patients. DESIGN.— We retrospectively analyzed the characteristics, MRD status, and prognosis of adult B-ALL based on a large real-world cohort of 486 patients during a 10-year period. RESULTS.— MRD assessed by MFC and polymerase chain reaction (PCR) assays for BCR-ABL+ patients showed concordant results in 74.2% of cases. MRD- status by our MFC panel could clearly predict a favorable relapse-free survival (RFS) and overall survival (OS) both at the end of induction and at the end of 1 consolidation course. Patients with continuous MRD- and with at least 1 MRD- result showed a favorable RFS and OS compared with those with at least 1 MRD+ result and continuous MRD+, respectively. CONCLUSIONS.— The single-tube 8-color MFC panel demonstrated a low cost, decent sensitivity, and comparability with polymerase chain reaction-MRD but an excellent performance in predicting RFS and OS, and thus could potentially be taken as a routine indicator in the evaluation of the treatment response for adult patients with B-ALL.
Collapse
Affiliation(s)
- Hongyan Liao
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Nenggang Jiang
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Ying Yang
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Xin Zhang
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jiao Chen
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Hongli Lai
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Qin Zheng
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Gao Q, Liu Y, Aypar U, Baik J, Londono D, Sun X, Zhang J, Zhang Y, Roshal M. Highly sensitive single tube B-lymphoblastic leukemia/lymphoma minimal/measurable residual disease test robust to surface antigen directed therapy. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:279-293. [PMID: 36999235 PMCID: PMC10508218 DOI: 10.1002/cyto.b.22120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/10/2023] [Accepted: 03/20/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND Measurement of minimal/measurable residual disease (MRD) in B-lymphoblastic leukemia/lymphoma (B-ALL) has become a routine clinical evaluation tool and remains the strongest predictor of treatment outcome. In recent years, new targeted anti-CD19 and anti-CD22 antibody-based and cellular therapies have revolutionized the treatment of the high-risk B-ALL. The new treatments raise challenges for diagnostic flow cytometry, which relies on the presence of specific surface antigens to identify the population of interest. So far, reported flow cytometry-based assays are developed to either achieve a deeper MRD level or to accommodate the loss of surface antigens post-target therapies, but not both. METHODS We developed a single tube flow cytometry assay (14-color-16-parameters). The method was validated using 94 clinical samples as well as spike-in and replicate experiments. RESULTS The assay was well suited for monitoring response to targeted therapies and reached a sensitivity below 10-5 with acceptable precision (coefficient of variation < 20%), accuracy, and interobserver variability (κ = 1). CONCLUSIONS The assay allows for sensitive disease detection of B-ALL MRD independent of CD19 and CD22 expression and allows uniform analysis of samples regardless of anti-CD19 and CD22 therapy.
Collapse
Affiliation(s)
- Qi Gao
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ying Liu
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Umut Aypar
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jeeyeon Baik
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dory Londono
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Xiaotian Sun
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jingping Zhang
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Yanming Zhang
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mikhail Roshal
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
16
|
Measurable residual disease in adult acute myeloid leukaemia: evaluation of a multidimensional 'radar' flow cytometric plot analysis method. Pathology 2023; 55:383-390. [PMID: 36725446 DOI: 10.1016/j.pathol.2022.10.007] [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: 04/04/2022] [Revised: 09/11/2022] [Accepted: 10/09/2022] [Indexed: 01/01/2023]
Abstract
Measurable residual disease (MRD) monitoring in acute myeloid leukaemia (AML) is becoming increasingly important and is predominantly performed by multiparameter flow cytometry (MFC) or quantitative polymerase chain reactions (RT-qPCR). We investigated the use of multidimensional plots (MD-MFC) for AML MRD monitoring in an adult cohort. AML MRD was determined using a novel MD-MFC method for 115 MRD samples. Results were correlated with traditional two-dimensional MFC (2D-MFC) and molecular methods. Using the standard cut-off of 0.1% CD45+ cells, concordance was 99/115 (p=0.332). Eighty-four of 115 were concordant using a very low reporting limit of 0.01% (p=0.216). MRD <0.1% by either method was present in 40 of 115 samples. Fifteen of 40 were MD-MFC positive and 2D-MFC negative. Of these two of 15 had a molecular MRD marker and both were positive. Molecular MRD markers were available in 36 of 115 cases. Twenty-one of 36 (58%) were concordant with MD-MFC. Eight of 36 had detectable molecular MRD only and eight of 36 had positive MD-MFC only. There was no correlation between either the MFC method and the molecular results. In summary, there is good correlation between MD- and 2D-MFC-MRD and no correlation between the MFC and molecular methods.
Collapse
|
17
|
Shimony S, Stahl M, Stone RM. Acute myeloid leukemia: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol 2023; 98:502-526. [PMID: 36594187 DOI: 10.1002/ajh.26822] [Citation(s) in RCA: 172] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023]
Abstract
DISEASE OVERVIEW Acute myeloid leukemia (AML) is a frequently fatal bone marrow stem cell cancer characterized by unbridled proliferation of malignant marrow stem cells with associated infection, anemia, and bleeding. An improved understanding of pathophysiology, improvements in measurement technology and at least 10 recently approved therapies have led to revamping the diagnostic, prognostic, and therapeutic landscape of AML. DIAGNOSIS One updated and one new classification system were published in 2022, both emphasizing the integration of molecular analysis into daily practice. Differences between the International Consensus Classification and major revisions from the previous 2016 WHO system provide both challenges and opportunities for care and clinical research. RISK ASSESSMENT AND MONITORING The European Leukemia Net 2022 risk classification integrates knowledge from novel molecular findings and recent trial results, as well as emphasizing dynamic risk based on serial measurable residual disease assessment. However, how to leverage our burgeoning ability to measure a small number of potentially malignant myeloid cells into therapeutic decision making is controversial. RISK ADAPTED THERAPY The diagnostic and therapeutic complexity plus the availability of newly approved agents requires a nuanced therapeutic algorithm which should integrate patient goals of care, comorbidities, and disease characteristics including the specific mutational profile of the patient's AML. The framework we suggest only represents the beginning of the discussion.
Collapse
Affiliation(s)
- Shai Shimony
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Rabin Medical Center and Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Meddi E, Savi A, Moretti F, Mallegni F, Palmieri R, Paterno G, Buzzatti E, Del Principe MI, Buccisano F, Venditti A, Maurillo L. Measurable Residual Disease (MRD) as a Surrogate Efficacy-Response Biomarker in AML. Int J Mol Sci 2023; 24:ijms24043062. [PMID: 36834477 PMCID: PMC9967250 DOI: 10.3390/ijms24043062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
In acute myeloid leukemia (AML) many patients experience relapse, despite the achievement of morphological complete remission; therefore, conventional morphologic criteria are currently considered inadequate for assessing the quality of the response after treatment. Quantification of measurable residual disease (MRD) has been established as a strong prognostic marker in AML and patients that test MRD negative have lower relapse rates and better survival than those who test positive. Different techniques, varying in their sensitivity and applicability to patients, are available for the measurement of MRD and their use as a guide for selecting the most optimal post-remission therapy is an area of active investigation. Although still controversial, MRD prognostic value promises to support drug development serving as a surrogate biomarker, potentially useful for accelerating the regulatory approval of new agents. In this review, we will critically examine the methods used to detect MRD and its potential role as a study endpoint.
Collapse
Affiliation(s)
- Elisa Meddi
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Arianna Savi
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Federico Moretti
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Flavia Mallegni
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Raffaele Palmieri
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | | | - Elisa Buzzatti
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | | | - Francesco Buccisano
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
- Correspondence:
| | - Luca Maurillo
- Hematology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
19
|
Chokr N, Gomez-Arteaga A. Measurable Residual Disease After CAR T-Cell Therapy. Semin Hematol 2023; 60:34-41. [PMID: 37080709 DOI: 10.1053/j.seminhematol.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/21/2023]
Abstract
Testing for measurable residual disease (MRD) provides important prognostic and predictive implications on survival and management of many hematologic diseases. Among the many clinical uses of MRD is post-therapy response assessment and risk stratification. With the integration of precision medicine in routine clinical care and the development of novel and innovative therapies resulting in deeper responses, it is necessary to refine the role of MRD, standardize available methodologies and define its role as a surrogate endpoint for relapse and time-to-next treatment in clinical studies. Chimeric Antigen Receptor (CAR) T-cell therapy is an approved treatment for various hematologic malignancies. Even though it produces high rates of remission, the durability of response is still a consideration as almost 40% to 50% of patients eventually relapse. MRD testing as a prognostic and surrogate marker is being explored in patients after CAR T-cell therapy to predict early relapse. In this chapter, we review the various tools available for MRD detection and monitoring post-CAR T-cell therapy. We later discuss disease-specific MRD assessment and its application in recent studies in the post-CAR T setting.
Collapse
|
20
|
Chen X, Gao Q, Roshal M, Cherian S. Flow cytometric assessment for minimal/measurable residual disease in B lymphoblastic leukemia/lymphoma in the era of immunotherapy. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:205-223. [PMID: 36683279 DOI: 10.1002/cyto.b.22113] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/30/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023]
Abstract
Minimal/measurable residual disease (MRD) is the most important independent prognostic factor for patients with B-lymphoblastic leukemia (B-LL). MRD post therapy has been incorporated into risk stratification and clinical management, resulting in substantially improved outcomes in pediatric and adult patients. Currently, MRD in B-ALL is most commonly assessed by multiparametric flow cytometry and molecular (polymerase chain reaction or high-throughput sequencing based) methods. The detection of MRD by flow cytometry in B-ALL often begins with B cell antigen-based gating strategies. Over the past several years, targeted immunotherapy directed against B cell markers has been introduced in patients with relapsed or refractory B-ALL and has demonstrated encouraging results. However, targeted therapies have significant impact on the immunophenotype of leukemic blasts, in particular, downregulation or loss of targeted antigens on blasts and normal B cell precursors, posing challenges for MRD detection using standard gating strategies. Novel flow cytometric approaches, using alternative strategies for population identification, sometimes including alternative gating reagents, have been developed and implemented to monitor MRD in the setting of post targeted therapy.
Collapse
Affiliation(s)
- Xueyan Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Qi Gao
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mikhail Roshal
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sindhu Cherian
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
21
|
Tecchio C, Russignan A, Krampera M. Immunophenotypic measurable residual disease monitoring in adult acute lymphoblastic leukemia patients undergoing allogeneic hematopoietic stem cell transplantation. Front Oncol 2023; 13:1047554. [PMID: 36910638 PMCID: PMC9992536 DOI: 10.3389/fonc.2023.1047554] [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: 09/18/2022] [Accepted: 01/11/2023] [Indexed: 02/24/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) offers a survival benefit to adult patients affected by acute lymphoblastic leukemia (ALL). However, to avoid an overt disease relapse, patients with pre or post transplant persistence or occurrence of measurable residual disease (MRD) may require cellular or pharmacological interventions with eventual side effects. While the significance of multiparametric flow cytometry (MFC) in the guidance of ALL treatment in both adult and pediatric patients is undebated, fewer data are available regarding the impact of MRD monitoring, as assessed by MFC analysis, in the allo-HSCT settings. Aim of this article is to summarize and discuss currently available information on the role of MFC detection of MRD in adult ALL patients undergoing allo-HSCT. The significance of MFC-based MRD according to sensitivity level, timing, and in relation to molecular techniques of MRD and chimerism assessment will be also discussed.
Collapse
Affiliation(s)
- Cristina Tecchio
- Department of Medicine, Section of Hematology and Bone Marrow Transplant Unit, University of Verona, Verona, Italy
| | - Anna Russignan
- Department of Medicine, Section of Hematology and Bone Marrow Transplant Unit, University of Verona, Verona, Italy
| | - Mauro Krampera
- Department of Medicine, Section of Hematology and Bone Marrow Transplant Unit, University of Verona, Verona, Italy
| |
Collapse
|
22
|
Paras G, Othus M, Schonhoff K, Shaw C, Sorror M, Halpern AB, Appelbaum J, Hendrie P, Walter RB, Estey EH, Percival MEM. Effect of ECOG performance status on outcomes in patients with acute myeloid leukemia and other high-grade myeloid neoplasms. Leukemia 2023; 37:231-234. [PMID: 36434064 PMCID: PMC9883154 DOI: 10.1038/s41375-022-01745-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Gabrielle Paras
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Megan Othus
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kelda Schonhoff
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Carole Shaw
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mohamed Sorror
- Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anna B Halpern
- Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jacob Appelbaum
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Paul Hendrie
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Roland B Walter
- Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Elihu H Estey
- Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mary-Elizabeth M Percival
- Department of Medicine, University of Washington, Seattle, WA, USA.
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
23
|
Allam S, Nasr K, Khalid F, Shah Z, Khan Suheb MZ, Mulla S, Vikash S, Bou Zerdan M, Anwer F, Chaulagain CP. Liquid biopsies and minimal residual disease in myeloid malignancies. Front Oncol 2023; 13:1164017. [PMID: 37213280 PMCID: PMC10196237 DOI: 10.3389/fonc.2023.1164017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/25/2023] [Indexed: 05/23/2023] Open
Abstract
Minimal residual disease (MRD) assessment through blood component sampling by liquid biopsies (LBs) is increasingly being investigated in myeloid malignancies. Blood components then undergo molecular analysis by flow cytometry or sequencing techniques and can be used as a powerful tool for prognostic and predictive purposes in myeloid malignancies. There is evidence and more is evolving about the quantification and identification of cell-based and gene-based biomarkers in myeloid malignancies to monitor treatment response. MRD based acute myeloid leukemia protocol and clinical trials are currently incorporating LB testing and preliminary results are encouraging for potential widespread use in clinic in the near future. MRD monitoring using LBs are not standard in myelodysplastic syndrome (MDS) but this is an area of active investigation. In the future, LBs can replace more invasive techniques such as bone marrow biopsies. However, the routine clinical application of these markers continues to be an issue due to lack of standardization and limited number of studies investigating their specificities. Integrating artificial intelligence (AI) could help simplify the complex interpretation of molecular testing and reduce errors related to operator dependency. Though the field is rapidly evolving, the applicability of MRD testing using LB is mostly limited to research setting at this time due to the need for validation, regulatory approval, payer coverage, and cost issues. This review focuses on the types of biomarkers, most recent research exploring MRD and LB in myeloid malignancies, ongoing clinical trials, and the future of LB in the setting of AI.
Collapse
Affiliation(s)
- Sabine Allam
- Department of Medicine and Medical Sciences, University of Balamand, Dekwaneh, Lebanon
| | - Kristina Nasr
- Department of Medicine and Medical Sciences, University of Balamand, Dekwaneh, Lebanon
| | - Farhan Khalid
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ, United States
| | - Zunairah Shah
- Department of Internal Medicine, Weiss Memorial Hospital, Chicago, IL, United States
| | | | - Sana Mulla
- Department of Internal Medicine, St Mary’s Medical Center, Apple Valley, CA, United States
| | - Sindhu Vikash
- Department of Medicine, Jacobi Medical center/AECOM Bronx, Bronx, NY, United States
| | - Maroun Bou Zerdan
- Department of Internal Medicine, SUNY Upstate Medical University, New York, NY, United States
| | - Faiz Anwer
- Department of Hematology and Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, United States
| | - Chakra P. Chaulagain
- Department of Hematology and Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, United States
- *Correspondence: Chakra P. Chaulagain,
| |
Collapse
|
24
|
Immunophenotype of Measurable Residual Blast Cells as an Additional Prognostic Factor in Adults with B-Cell Acute Lymphoblastic Leukemia. Diagnostics (Basel) 2022; 13:diagnostics13010021. [PMID: 36611312 PMCID: PMC9818326 DOI: 10.3390/diagnostics13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Measurable residual disease (MRD) is a well-known independent prognostic factor in acute leukemias, and multicolor flow cytometry (MFC) is widely used to detect MRD. MFC is able not only to enumerate MRD accurately but also to describe an antigen expression profile of residual blast cells. However, the relationship between MRD immunophenotype and patient survival probability has not yet been studied. We determined the prognostic impact of MRD immunophenotype in adults with B-cell acute lymphoblastic leukemia (B-ALL). In a multicenter study RALL-2016 (NCT03462095), 267 patients were enrolled from 2016 to 2022. MRD was assessed at the end of induction (day 70) in 94 patients with B-ALL by six- or 10-color flow cytometry in the bone marrow specimens. The 4 year relapse-free survival (RFS) was lower in MRD-positive B-ALL patients [37% vs. 78% (p < 0.0001)]. The absence of CD10, positive expression of CD38, and high expression of CD58 on MRD cells worsened the 4 year RFS [19% vs. 51% (p = 0.004), 0% vs. 51% (p < 0.0001), and 21% vs. 40% (p = 0.02), respectively]. The MRD immunophenotype is associated with RFS and could be an additional prognostic factor for B-ALL patients.
Collapse
|
25
|
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
|
26
|
Glencross DK, Swart L, Pretorius M, Lawrie D. Evaluation of fixed-panel, multicolour ClearLLab 10C at an academic flow cytometry laboratory in Johannesburg, South Africa. Afr J Lab Med 2022; 11:1458. [PMID: 35937760 PMCID: PMC9350555 DOI: 10.4102/ajlm.v11i1.1458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Background Flow cytometric immunophenotyping is well established for the diagnosis of haematological neoplasms. New commercially available systems offer fixed, pre-aliquoted multi-parameter analysis to simplify sample preparation and standardise data analysis. Objective The Beckman Coulter (BC) ClearLLab™ 10C (4-tube) system was evaluated against an existing laboratory developed test (LDT). Methods Peripheral blood and bone marrow aspirates (n = 101), tested between August 2019 and November 2019 at an academic pathology laboratory in Johannesburg, South Africa, were analysed. Following daily instrument quality control, samples were prepared for LDT (using > 20 2–4-colour in-house panels and an extensive liquid monoclonal reagent repertoire) or ClearLLab 10C, and respectively analysed using in-house protocols on a Becton Dickinson FACSCalibur, or manufacturer-directed protocols on a BC Navios. Becton Dickinson Paint-a-Gate or BC Kaluza C software facilitated data interpretation. Diagnostic accuracy (concordance) was established by calculating sensitivity and specificity outcomes. Results Excellent agreement (clinical diagnostic concordance) with 100% specificity and sensitivity was established between LDT and ClearLLab 10C in 67 patients with a haematological neoplasm and 34 participants with no haematological disease. Similar acceptable diagnostic concordance (97%) was noted when comparing ClearLLab 10C to clinicopathological outcomes. Additionally, the ClearLLab 10C panels, analysed with Kaluza C software, enabled simultaneous discrimination of disease and concurrent background myeloid and lymphoid haematological populations, including assessing stages of maturation or sub-populations. Conclusion ClearLLab 10C panels provide excellent agreement to existing LDTs and may reliably be used for immunophenotyping of haematological neoplasms, simplifying and standardising sample preparation and data acquisition.
Collapse
Affiliation(s)
- Deborah K Glencross
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, Charlotte Maxeke Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| | - Leanne Swart
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, Charlotte Maxeke Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| | - Melanie Pretorius
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, Charlotte Maxeke Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| | - Denise Lawrie
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, Charlotte Maxeke Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
27
|
Feng J, Guo Y, Yang W, Zou Y, Zhang L, Chen Y, Zhang Y, Zhu X, Chen X. Childhood Acute B-Lineage Lymphoblastic Leukemia With CDKN2A/B Deletion Is a Distinct Entity With Adverse Genetic Features and Poor Clinical Outcomes. Front Oncol 2022; 12:878098. [PMID: 35712467 PMCID: PMC9195293 DOI: 10.3389/fonc.2022.878098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
To further emphasize the clinical-genetic features and prognosis of CDKN2A/B deletions in childhood acute lymphoblastic leukemia (ALL), we retrospectively analyzed 819 consecutive B-ALL patients treated with the Chinese Children's Cancer Group ALL-2015 (CCCG-ALL-2015) protocol, and fluorescence in situ hybridization (FISH) analysis on CDKN2A/B deletion was available for 599 patients. The prevalence of CDKN2A/B gene deletions was 20.2% (121/599) of B-ALL. CDKN2A/B deletions were significantly associated with older age, higher leukocyte counts, a higher percentage of hepatosplenomegaly, and a higher frequency of BCR-ABL (p < 0.05). Those patients achieved similar minimal residual disease (MRD) clearance and complete remission compared to patients without CDKN2A/B deletion. The CDKN2A/B deletions were correlated with inferior outcomes, including a 3-year event-free survival (EFS) rate (69.8 ± 4.6 vs. 89.2 ± 1.6%, p = 0.000) and a 3-year overall survival (OS) rate (89.4% ± 2.9% vs. 94.7% ± 1.1%, p = 0.037). In multivariable analysis, CDKN2A/B deletion was still an independent prognostic factor for EFS in total cohorts (p < 0.05). We also detected a multiplicative interaction between CDKN2A/B deletions and TP53 deletion on dismal prognosis (p-interaction < 0.05). In conclusion, CDKN2A/B deletion is associated with distinct characteristics and serves as a poor prognostic factor in pediatric ALL, especially in TP53 deletion carriers.
Collapse
Affiliation(s)
- Jing 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
| | - Ye Guo
- 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
| | - Wenyu Yang
- 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
| | - Yao Zou
- 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
| | - Li 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
| | - Yumei 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
| | - Yingchi 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
| | - Xiaofan Zhu
- 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
| | - Xiaojuan 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
| |
Collapse
|
28
|
Hein K, Short N, Jabbour E, Yilmaz M. Clinical Value of Measurable Residual Disease in Acute Lymphoblastic Leukemia. Blood Lymphat Cancer 2022; 12:7-16. [PMID: 35340663 PMCID: PMC8943430 DOI: 10.2147/blctt.s270134] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 02/24/2022] [Indexed: 01/04/2023]
Abstract
Measurable (minimal) residual disease (MRD) status in acute lymphoblastic leukemia (ALL) has largely superseded the importance of traditional risk factors for ALL, such as baseline white blood cell count, cytogenetics, and immunophenotype, and has emerged as the most powerful independent prognostic predictor. The development of sensitive MRD techniques, such as multicolor flow cytometry (MFC), quantitative polymerase chain reaction (PCR), and next-generation sequencing (NGS), may further improve risk stratification and expand its impact in therapy. Additionally, the availability of highly effective agents for MRD eradication, such as blinatumomab, inotuzumab ozogamicin, and chimeric antigen receptor (CAR) T-cell therapies, enabled the development of frontline regimens capable of eradicating MRD early in the treatment course. While long-term follow-up of this approach is lacking, it has the potential to significantly reduce the need for intensive post-remission treatments, including allogeneic bone marrow transplantation, in a significant proportion of patients with ALL.
Collapse
Affiliation(s)
- Kyaw Hein
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
29
|
Buccisano F, Palmieri R, Piciocchi A, Arena V, Maurillo L, Del Principe MI, Paterno G, Irno-Consalvo MA, Ottone T, Divona M, Conti C, Fraboni D, Lavorgna S, Arcese W, Voso MT, Venditti A. Clinical relevance of an objective flow cytometry approach based on limit of detection and limit of quantification for measurable residual disease assessment in acute myeloid leukemia. A post-hoc analysis of the GIMEMA AML1310 trial. Haematologica 2022; 107:2823-2833. [PMID: 35295076 PMCID: PMC9713557 DOI: 10.3324/haematol.2021.279777] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Using a multiparametric flow cytometry assay, we assessed the predictive power of a threshold calculated applying the criteria of limit of detection (LOD) and limit of quantitation (LOQ) in adult patients with acute myeloid leukemia. This was a post-hoc analysis of 261 patients enrolled in the GIMEMA AML1310 prospective trial. According to the protocol design, using the predefined measurable residual disease (MRD) threshold of 0.035% bone marrow residual leukemic cells (RLC) calculated on mononuclear cells, 154 (59%) of the 261 patients were negative (MRD <0.035%) and 107 (41%) were positive (MRD ≥0.035%). Using LOD and LOQ, we selected the following categories of patients: (i) LODneg if RLC were below the LOD (74; 28.4%); (ii) LODpos-LOQneg if RLC were between the LOD and LOQ (43; 16.5%); and (iii) LOQpos if RLC were above the LOQ (144; 54.4%). Two-year overall survival of these three categories of patients was 75.4%, 79.8% and 66.4%, respectively (P=0.1197). Given their superimposable outcomes, the LODneg and LODpos-LOQneg categories were combined. Two-year overall survival of LODneg/LODpos-LOQneg patients was 77.0% versus 66.4% of LOQpos individuals (P=0.043). This figure was challenged in univariate analysis (P=0.046, hazard ratio=1.6, 95% confidence interval: 1.01-2.54) which confirmed the independent role of the LOD-LOQ approach in determining overall survival. In the AML1310 protocol, using the threshold of 0.035%, 2-year overall survival of patients with MRD <0.035% and MRD ≥0.035% was 74.5% versus 66.4%, respectively (P=0.3521). In conclusion, the use of the LOD-LOQ method results in more sensitive detection of MRD that, in turn, translates into a more accurate recognition of patients with different outcomes.
Collapse
Affiliation(s)
- Francesco Buccisano
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,FB and RP contributed equally as co-first authors
| | - Raffaele Palmieri
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,FB and RP contributed equally as co-first authors
| | | | | | - Luca Maurillo
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | | | | | | | - Tiziana Ottone
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Mariadomenica Divona
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Consuelo Conti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Daniela Fraboni
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Serena Lavorgna
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - William Arcese
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,Rome Transplant Network, Rome, Italy
| | - Maria Teresa Voso
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Adriano Venditti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| |
Collapse
|
30
|
Singh J, Gorniak M, Grigoriadis G, Westerman D, McBean M, Venn N, Law T, Sutton R, Morgan S, Fleming S. Correlation between a 10-color flow cytometric measurable residual disease (MRD) analysis and molecular MRD in adult B-acute lymphoblastic leukemia. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2022; 102:115-122. [PMID: 34806309 DOI: 10.1002/cyto.b.22043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/21/2021] [Accepted: 11/10/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND Measurable residual disease (MRD) monitoring in acute lymphoblastic leukemia (ALL) is an important predictive factor for patient outcome and treatment intensification. Molecular monitoring, particularly with quantitative polymerase chain reaction (qPCR) to measure immunoglobin heavy or kappa chain (Ig) or T-cell receptor (TCR) gene rearrangements, offers high sensitivity but accessibility is limited by expertise, cost, and turnaround time. Flow cytometric assays are cheaper and more widely available, and sensitivity is improved with multi-parameter flow cytometry at eight or more colors. METHODS We developed a 10-color single tube flow cytometry assay. Samples were subject to bulk ammonium chloride lysis to maximize cell yields with a target of 1 × 106 events. Once normal maturation patterns were established, patient samples were analyzed in parallel to standard molecular monitoring. RESULTS Flow cytometry was performed on 114 samples. An informative immunophenotype was identifiable in all 22 patients who had a diagnostic sample. MRD analysis was performed on 87 samples. The median lower limits of detection and quantification were 0.004% (range 0.0005%-0.028%) and 0.01% (range 0.001%-0.07%) respectively. Sixty-five samples had concurrent molecular MRD testing, with good correlation (r = 0.83, p < 0.001). Results were concordant in 52 samples, and discordant in 13 samples, including one case where impending relapse was detected by flow cytometry but not Ig/TCR qPCR. CONCLUSIONS Our 10-color flow cytometric MRD assay provided adequate sensitivity and good correlation with molecular assays. This technique offers rapid and affordable testing in B-ALL patients, including cases where a suitable molecular assay cannot be developed or has reduced sensitivity.
Collapse
Affiliation(s)
- Jasmine Singh
- Laboratory Haematology, Alfred Pathology, Melbourne, Victoria, Australia
| | - Malgorzata Gorniak
- Laboratory Haematology, Alfred Pathology, Melbourne, Victoria, Australia
| | - George Grigoriadis
- Laboratory Haematology, Alfred Pathology, Melbourne, Victoria, Australia.,Clinical Haematology, Monash Health, Clayton, Victoria, Australia
| | - David Westerman
- Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michelle McBean
- Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Nicola Venn
- Children's Cancer Institute and School of Women's and Children's Health, UNSW, Sydney, New South Wales, Australia
| | - Tamara Law
- Children's Cancer Institute and School of Women's and Children's Health, UNSW, Sydney, New South Wales, Australia
| | - Rosemary Sutton
- Children's Cancer Institute and School of Women's and Children's Health, UNSW, Sydney, New South Wales, Australia
| | - Sue Morgan
- Laboratory Haematology, Alfred Pathology, Melbourne, Victoria, Australia
| | - Shaun Fleming
- Laboratory Haematology, Alfred Pathology, Melbourne, Victoria, Australia.,Clinical Haematology, Monash Health, Clayton, Victoria, Australia
| |
Collapse
|
31
|
"Blasts" in myeloid neoplasms - how do we define blasts and how do we incorporate them into diagnostic schema moving forward? Leukemia 2022; 36:327-332. [PMID: 35042955 DOI: 10.1038/s41375-021-01498-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 11/08/2022]
|
32
|
Technical Aspects of Flow Cytometry-based Measurable Residual Disease Quantification in Acute Myeloid Leukemia: Experience of the European LeukemiaNet MRD Working Party. Hemasphere 2022; 6:e676. [PMID: 34964040 PMCID: PMC8701786 DOI: 10.1097/hs9.0000000000000676] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Measurable residual disease (MRD) quantified by multiparameter flow cytometry (MFC) is a strong and independent prognostic factor in acute myeloid leukemia (AML). However, several technical factors may affect the final read-out of the assay. Experts from the MRD Working Party of the European LeukemiaNet evaluated which aspects are crucial for accurate MFC-MRD measurement. Here, we report on the agreement, obtained via a combination of a cross-sectional questionnaire, live discussions, and a Delphi poll. The recommendations consist of several key issues from bone marrow sampling to final laboratory reporting to ensure quality and reproducibility of results. Furthermore, the experiences were tested by comparing two 8-color MRD panels in multiple laboratories. The results presented here underscore the feasibility and the utility of a harmonized theoretical and practical MFC-MRD assessment and are a next step toward further harmonization.
Collapse
|
33
|
Salhotra A, Tsai NC, Zhang J, Ngo D, Aribi A, Sandhu K, Ball B, Al-Malki M, Ali H, Koller P, Artz A, Forman S, Nakamura R, Stein A, Marcucci G, Aldoss I, Pullarkat V. Venetoclax and hypomethylating agents yield high response rates and favourable transplant outcomes in patients with newly diagnosed acute myeloid leukaemia. Br J Haematol 2021; 196:e71-e74. [PMID: 34931310 DOI: 10.1111/bjh.17996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Amandeep Salhotra
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Ni-Chun Tsai
- Department of Information Sciences, City of Hope National Medical Center, Duarte, CA, USA
| | - Jianying Zhang
- Department of Information Sciences, City of Hope National Medical Center, Duarte, CA, USA
| | - Dat Ngo
- Department of Clinical Pharmacology, City of Hope National Medical Center, Duarte, CA, USA
| | - Ahmed Aribi
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Karamjeet Sandhu
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Brian Ball
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Monzr Al-Malki
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Haris Ali
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Paul Koller
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Andrew Artz
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Stephen Forman
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Ryotaro Nakamura
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Anthony Stein
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Guido Marcucci
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Ibrahim Aldoss
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| | - Vinod Pullarkat
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
34
|
Arumugam JR, Bommannan K, Radhakrishnan V, Sagar TG, Sundersingh S. Immunophenotypic expression and immunomodulation in minimal residual disease analysis of pediatric B acute lymphoblastic leukemia by high sensitive flow cytometry. Leuk Lymphoma 2021; 63:644-652. [PMID: 34727819 DOI: 10.1080/10428194.2021.1992755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The major challenge in minimal residual disease (MRD) detection is the antigen modulation in post treated samples restraining the use of diagnostic immunophenotypic (IP) signature of leukemic blasts for MRD detection. The IP expression of 10 antigens in 167 children diagnosed as B-acute lymphoblastic leukemia (B-ALL) in comparison to hematogones and the extent of immunomodulation in 60 post treated MRD positive cases were studied. Upregulation was the predictable shift noted in antigens like CD73, CD86, CD19, CD20 and CD45 which was statistically significant for all except CD45. Downregulation was the predictable shift noted in antigens like CD10, CD38, CD58 and CD34 and was statistically significant in all. CD123 showed no significant trend. This immunomodulation in B-ALL results in aberrant expression of antigens during follow-up compared to the diagnostic phenotypic pattern. Hence it is necessary to be aware of the immunomodulations of antigens used in primary diagnosis to avoid being misled during MRD analysis.
Collapse
Affiliation(s)
- Jhansi Rani Arumugam
- Departments of Oncopathology, Cancer Institute (Women's India Association), Chennai, India
| | - Karthik Bommannan
- Departments of Oncopathology, Cancer Institute (Women's India Association), Chennai, India
| | | | - Tenali Gnana Sagar
- Departments of Oncopathology, Cancer Institute (Women's India Association), Chennai, India
| | - Shirley Sundersingh
- Departments of Oncopathology, Cancer Institute (Women's India Association), Chennai, India
| |
Collapse
|
35
|
The role of CD44 in the assessment of minimal residual disease of multiple myeloma by flow cytometry. J Hematop 2021. [DOI: 10.1007/s12308-021-00468-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
36
|
Pediatric Mixed-Phenotype Acute Leukemia: What's New? Cancers (Basel) 2021; 13:cancers13184658. [PMID: 34572885 PMCID: PMC8469808 DOI: 10.3390/cancers13184658] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Pediatric mixed-phenotype leukemia is a rare form of blood cancer in children. In this review, we cover both the evolution of treatment over the past several years and outline new emerging concepts in this disease. Abstract Mixed-phenotype acute leukemias (MPAL) are rare in children and often lack consensus on optimal management. This review examines the current controversies and emerging paradigms in the management of pediatric MPAL. We examine risk stratification, outcomes of recent retrospective and prospective collaborative trials, and the role of transplantation and precision genomics, and outline emerging targets and concepts in this rare entity.
Collapse
|
37
|
Chatterjee G, Sriram H, Ghogale S, Deshpande N, Khanka T, Girase K, Verma S, Arolkar G, Dasgupta N, Narula G, Shetty D, Dhamne C, Moulik NR, Rajpal S, Patkar NV, Banavali S, Gujral S, Subramanian PG, Tembhare PR. Mimics and artefacts of measurable residual disease in a highly sensitive multicolour flow cytometry assay for B-lymphoblastic leukaemia/lymphoma: critical consideration for analysis of measurable residual disease. Br J Haematol 2021; 196:374-379. [PMID: 34476808 DOI: 10.1111/bjh.17801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/13/2021] [Indexed: 01/08/2023]
Abstract
High-sensitivity multicolour flow cytometry (MFC)-based B-lymphoblastic leukaemia (B-ALL) measurable residual disease (BMRD) assay is increasingly being used in clinical practice. Herein, we describe six consistently present low-level populations immunophenotypically mimicking abnormal B-ALL blasts in 441 BMRD samples from 301 children. These included CD19+ CD123+ plasmacytoid dendritic cells differentiating from lymphoid precursors, CD10+ transitional B cells with CD10+ /CD38dim-to-negative/CD20bright/CD45bright phenotype, CD19+ natural killer (NK) cells, CD73bright/CD10+ mesenchymal stromal/stem cells, CD73bright/CD34+ endothelial cells, and a CD34+ CD38dim-to-negative/CD10- /CD20bright/CD45bright subset of mature B cells. We provide the proportions, comprehensive immunophenotype, and practical clues for proper identification of these low-level populations. Knowledge regarding the presence and immunophenotype of these mimics is essential for accurate interpretation in high-sensitivity MFC-BMRD analysis.
Collapse
Affiliation(s)
- Gaurav Chatterjee
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Harshini Sriram
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Sitaram Ghogale
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Nilesh Deshpande
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Twinkle Khanka
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Karishma Girase
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Shefali Verma
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Gauri Arolkar
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Niharika Dasgupta
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Gaurav Narula
- Department of Pediatric Oncology, Tata Memorial Hospital, Tata Memorial Centre, HBNI University, Mumbai, India
| | - Dhanalaxmi Shetty
- Department of Cancer Cytogenetics, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Chetan Dhamne
- Department of Pediatric Oncology, Tata Memorial Hospital, Tata Memorial Centre, HBNI University, Mumbai, India
| | - Nirmalya R Moulik
- Department of Pediatric Oncology, Tata Memorial Hospital, Tata Memorial Centre, HBNI University, Mumbai, India
| | - Sweta Rajpal
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Nikhil V Patkar
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Shripad Banavali
- Department of Pediatric Oncology, Tata Memorial Hospital, Tata Memorial Centre, HBNI University, Mumbai, India
| | - Sumeet Gujral
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Papagudi G Subramanian
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| | - Prashant R Tembhare
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, HBNI University, Navi Mumbai, India
| |
Collapse
|
38
|
Abstract
Minimal or measurable residual disease (MRD) after therapy is the most important independent prognostic factor in acute myeloid leukemia. MRD measured by multiparametric flow cytometry and real-time quantitative polymerase chain reaction has been integrated into risk stratification and used to guide future treatment strategies. Recent technological advances have allowed the application of the novel molecular method, high-throughput sequencing, in MRD detection in clinical practice to improve sensitivity and specificity. Randomized studies are needed to address outstanding issues, including the optimal methods and timing of MRD testing and interlaboratory standardization to facilitate comparisons, to further improve MRD-directed interventions.
Collapse
Affiliation(s)
- Xueyan Chen
- Hematopathology, SCCA G7800, 825 Eastlake Ave E., Seattle, WA 98109, USA
| | - Sindhu Cherian
- Hematopathology, SCCA G7800, 825 Eastlake Ave E., Seattle, WA 98109, USA.
| |
Collapse
|
39
|
Rolf N, Liu LYT, Tsang A, Lange PF, Lim CJ, Maxwell CA, Vercauteren SM, Reid GSD. A cross-standardized flow cytometry platform to assess phenotypic stability in precursor B-cell acute lymphoblastic leukemia (B-ALL) xenografts. Cytometry A 2021; 101:57-71. [PMID: 34128309 PMCID: PMC9292200 DOI: 10.1002/cyto.a.24473] [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: 03/02/2021] [Revised: 05/07/2021] [Accepted: 06/10/2021] [Indexed: 11/17/2022]
Abstract
With the continued poor outcome of relapsed acute lymphoblastic leukemia (ALL), new patient‐specific approaches for disease progression monitoring and therapeutic intervention are urgently needed. Patient‐derived xenografts (PDX) of primary ALL in immune‐deficient mice have become a powerful tool for studying leukemia biology and therapy response. In PDX mice, the immunophenotype of the patient's leukemia is commonly believed to be stably propagated. In patients, however, the surface marker expression profile of the leukemic population often displays poorly understood immunophenotypic shifts during chemotherapy and ALL progression. We therefore developed a translational flow cytometry platform to study whether the patient‐specific immunophenotype is faithfully recapitulated in PDX mice. To enable valid assessment of immunophenotypic stability and subpopulation complexity of the patient's leukemia after xenotransplantation, we comprehensively immunophenotyped diagnostic B‐ALL from children and their matched PDX using identical, clinically standardized flow protocols and instrument settings. This cross‐standardized approach ensured longitudinal stability and cross‐platform comparability of marker expression intensity at high phenotyping depth. This analysis revealed readily detectable changes to the patient leukemia‐associated immunophenotype (LAIP) after xenotransplantation. To further investigate the mechanism underlying these complex immunophenotypic shifts, we applied an integrated analytical approach that combined clinical phenotyping depth and high analytical sensitivity with unbiased high‐dimensional algorithm‐based analysis. This high‐resolution analysis revealed that xenotransplantation achieves patient‐specific propagation of phenotypically stable B‐ALL subpopulations and that the immunophenotypic shifts observed at the level of bulk leukemia were consistent with changes in underlying subpopulation abundance. By incorporating the immunophenotypic complexity of leukemic populations, this novel cross‐standardized analytical platform could greatly expand the utility of PDX for investigating ALL progression biology and assessing therapies directed at eliminating relapse‐driving leukemic subpopulations.
Collapse
Affiliation(s)
- Nina Rolf
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lorraine Y T Liu
- Clinical Immunology Lab, Division of Hematopathology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Angela Tsang
- Clinical Immunology Lab, Division of Hematopathology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Philipp F Lange
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chinten James Lim
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher A Maxwell
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suzanne M Vercauteren
- Clinical Immunology Lab, Division of Hematopathology, BC Children's Hospital, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gregor S D Reid
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
40
|
Roerden M, Wirths S, Sökler M, Bethge WA, Vogel W, Walz JS. Impact of Mantle Cell Lymphoma Contamination of Autologous Stem Cell Grafts on Outcome after High-Dose Chemotherapy. Cancers (Basel) 2021; 13:cancers13112558. [PMID: 34071000 PMCID: PMC8197101 DOI: 10.3390/cancers13112558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 11/19/2022] Open
Abstract
Simple Summary High-dose chemotherapy followed by autologous hematopoietic stem cell transplantation (Auto-HSCT) is a standard frontline treatment for fit mantle cell lymphoma (MCL) patients. As there is a need for predictive factors to identify patients unlikely to benefit from this therapy, we investigated the prognostic impact of lymphoma cell contamination of autologous stem cell grafts. Analyzing a cohort of 36 MCL patients, we show that lymphoma cell contamination of stem cell grafts is associated with poor outcomes after Auto-HSCT. Its analysis might thus improve risk assessment and enable risk-stratified treatment strategies for MCL patients. Abstract Novel predictive factors are needed to identify mantle cell lymphoma (MCL) patients at increased risk for relapse after high-dose chemotherapy and autologous hematopoietic stem cell transplantation (HDCT/Auto-HSCT). Although bone marrow and peripheral blood involvement is commonly observed in MCL and lymphoma cell contamination of autologous stem cell grafts might facilitate relapse after Auto-HSCT, prevalence and prognostic significance of residual MCL cells in autologous grafts are unknown. We therefore performed a multiparameter flow cytometry (MFC)-based measurable residual disease (MRD) assessment in autologous stem cell grafts and analyzed its association with clinical outcome in an unselected retrospective cohort of 36 MCL patients. MRD was detectable in four (11%) autologous grafts, with MRD levels ranging from 0.002% to 0.2%. Positive graft-MRD was associated with a significantly shorter progression-free and overall survival when compared to graft-MRD negative patients (median 9 vs. 56 months and 25 vs. 132 months, respectively) and predicted early relapse after Auto-HSCT (median time to relapse 9 vs. 44 months). As a predictor of outcome after HDCT/Auto-HSCT, MFC-based assessment of graft-MRD might improve risk stratification and support clinical decision making for risk-oriented treatment strategies in MCL.
Collapse
Affiliation(s)
- Malte Roerden
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Correspondence:
| | - Stefan Wirths
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Martin Sökler
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Wolfgang A. Bethge
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Wichard Vogel
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Juliane S. Walz
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology (IKP) and Robert Bosch Center for Tumor Diseases (RBCT), Auerbachstr. 112, 70376 Stuttgart, Germany
| |
Collapse
|
41
|
Consensus Recommendations for MRD Testing in Adult B-Cell Acute Lymphoblastic Leukemia in Ontario. ACTA ACUST UNITED AC 2021; 28:1376-1387. [PMID: 33808300 PMCID: PMC8025812 DOI: 10.3390/curroncol28020131] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022]
Abstract
Measurable (minimal) residual disease (MRD) is an established, key prognostic factor in adult B-cell acute lymphoblastic leukemia (B-ALL), and testing for MRD is known to be an important tool to help guide treatment decisions. The clinical value of MRD testing depends on the accuracy and reliability of results. Currently, there are no Canadian provincial or national guidelines for MRD testing in adult B-ALL, and consistent with the absence of such guidelines, there is no uniform Ontario MRD testing consensus. Moreover, there is great variability in Ontario in MRD testing with respect to where, when, and by which technique, MRD testing is performed, as well as in how the results are interpreted. To address these deficiencies, an expert multidisciplinary working group was convened to define consensus recommendations for improving the provision of such testing. The expert panel recommends that MRD testing should be implemented in a centralized manner to ensure expertise and accuracy in testing for this low volume indication, thereby to provide accurate, reliable results to clinicians and patients. All adult patients with B-ALL should receive MRD testing after induction chemotherapy. Philadelphia chromosome (Ph)-positive patients should have ongoing monitoring of MRD during treatment and thereafter, while samples from Ph-negative B-ALL patients should be tested at least once later during treatment, ideally at 12 to 16 weeks after treatment initiation. In Ph-negative adult B-ALL patients, standardized, ideally centralized, protocols must be used for MRD testing, including both flow cytometry and immunoglobulin (Ig) heavy chain and T-cell receptor (TCR) gene rearrangement analysis. For Ph-positive B-ALL patients, MRD testing using a standardized protocol for reverse transcription real-time quantitative PCR (RT-qPCR) for the BCR-ABL1 gene fusion transcript is recommended, with Ig/TCR gene rearrangement analysis done in parallel likely providing additional clinical information.
Collapse
|
42
|
Seth N, Mahajan V, Kedia S, Sutar A, Sehgal K. Minimal Residual Disease (MRD) detection in B- ALL – Experience of a standalone flow cytometry laboratory. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2021. [DOI: 10.1016/j.phoj.2020.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
43
|
Liu Z, Li Y, Shi C. Monitoring minimal/measurable residual disease in B-cell acute lymphoblastic leukemia by flow cytometry during targeted therapy. Int J Hematol 2021; 113:337-343. [PMID: 33502735 DOI: 10.1007/s12185-021-03085-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 11/24/2022]
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is a hematologic malignancy of B-type lymphoid precursor cells. Minimal/measurable residual disease (MRD) is an important prognostic factor for B-ALL relapse. Traditional flow cytometry detection mainly relies on CD19-based gating strategies. However, relapse of CD19-negative B-ALL frequently occurs in patients who receive cellular and targeted therapy. This review will summarize the technical aspects of standard MRD assessment in B-ALL by flow cytometry, and then discuss the challenges of MRD strategies to deal with the scenario of CD19 negative or dim B-ALL relapse.
Collapse
Affiliation(s)
- Zhiyu Liu
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Li
- Central Laboratory of Hematology and Oncology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ce Shi
- Central Laboratory of Hematology and Oncology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
44
|
Chatterjee G, Dudakia V, Ghogale S, Deshpande N, Girase K, Chaturvedi A, Shetty D, Senger M, Jain H, Bagal B, Bonda A, Punatar S, Gokarn A, Khattry N, Patkar NV, Gujral S, Subramanian PG, Tembhare PR. Expression of CD304/neuropilin-1 in adult b-cell lymphoblastic leukemia/lymphoma and its utility for the measurable residual disease assessment. Int J Lab Hematol 2021; 43:990-999. [PMID: 33432783 DOI: 10.1111/ijlh.13456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/27/2020] [Accepted: 12/14/2020] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Many new markers are being evaluated to increase the sensitivity and applicability of multicolor flow cytometry (MFC)-based measurable residual disease (MRD) monitoring. However, most of the studies are limited to childhood B-cell lymphoblastic leukemia/lymphoma (B-ALL), and reports in adult B-ALL are extremely scarce and limited to small cohorts. We studied the expression of CD304/neuropilin-1 in a large cohort of adult B-ALL patients and evaluated its practical utility in MFC-based MRD analysis. METHODS CD304 was studied in blasts from adult B-ALL patients and normal precursor B cells (NPBC) from non-B-ALL bone marrow samples using MFC. CD304 expression intensity and pattern were studied with normalized-mean fluorescent intensity (nMFI) and coefficient of variation of immunofluorescence (CVIF), respectively. MFC-based MRD was performed at end of induction (EOI; day-35), end of consolidation (EOC; day 78-80), and subsequent follow-up (SFU) time points. RESULTS CD304 was positive in 120/214(56.07%) and was significantly associated with BCR-ABL1 fusion (P = .001). EOI-MRD and EOC-MRD were positive in 129/214(60.3%) and 50/81(61.72%), respectively. CD304 was positive in a significant percentage of EOI (48%, 62/129) and EOC (52%, 26/50) MRD-positive B-ALL samples. Its expression was retained, lost, and gained in 73.7%, 26.3%, and 11.3% of EOI-MRD and 85.7%, 14.3%, and none of EOC-MRD samples, respectively. Low-level MRD (<0.01%) was detectable in 34 of all (EOI + EOC + SFU = 189) MRD-positive samples, and CD304 was found useful in 50% of these samples. CONCLUSION CD304 is commonly expressed in adult B-ALL and clearly distinguish B-ALL blasts from normal precursor B cells. It is a stable MRD marker and distinctly useful in the detection of MFC-based MRD monitoring, especially in high-sensitivity MRD assay.
Collapse
Affiliation(s)
- Gaurav Chatterjee
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Vishesh Dudakia
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Sitaram Ghogale
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Nilesh Deshpande
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Karishma Girase
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Anumeha Chaturvedi
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Dhanlaxmi Shetty
- Department of Department of Cancer Cytogenetics, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Manju Senger
- Department of Medical Oncology, Tata Memorial Center, HBNI University, Mumbai, India
| | - Hasmukh Jain
- Department of Medical Oncology, Tata Memorial Center, HBNI University, Mumbai, India
| | - Bhausaheb Bagal
- Department of Medical Oncology, Tata Memorial Center, HBNI University, Mumbai, India
| | - Avinash Bonda
- Department of Medical Oncology, Tata Memorial Center, HBNI University, Mumbai, India
| | - Sachin Punatar
- Department of Medical Oncology, Tata Memorial Center, HBNI University, Mumbai, India
| | - Anant Gokarn
- Department of Medical Oncology, Tata Memorial Center, HBNI University, Mumbai, India
| | - Navin Khattry
- Department of Medical Oncology, Tata Memorial Center, HBNI University, Mumbai, India
| | - Nikhil V Patkar
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Sumeet Gujral
- Department of Pathology, Tata Memorial Center, HBNI University, Mumbai, India
| | - Papagudi G Subramanian
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Prashant R Tembhare
- Department of Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| |
Collapse
|
45
|
Kadauke S, Myers RM, Li Y, Aplenc R, Baniewicz D, Barrett DM, Barz Leahy A, Callahan C, Dolan JG, Fitzgerald JC, Gladney W, Lacey SF, Liu H, Maude SL, McGuire R, Motley LS, Teachey DT, Wertheim GB, Wray L, DiNofia AM, Grupp SA. Risk-Adapted Preemptive Tocilizumab to Prevent Severe Cytokine Release Syndrome After CTL019 for Pediatric B-Cell Acute Lymphoblastic Leukemia: A Prospective Clinical Trial. J Clin Oncol 2021; 39:920-930. [PMID: 33417474 DOI: 10.1200/jco.20.02477] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
PURPOSE To prospectively evaluate the effectiveness of risk-adapted preemptive tocilizumab (PT) administration in preventing severe cytokine release syndrome (CRS) after CTL019, a CD19 chimeric antigen receptor T-cell therapy. METHODS Children and young adults with CD19-positive relapsed or refractory B-cell acute lymphoblastic leukemia were assigned to high- (≥ 40%) or low- (< 40%) tumor burden cohorts (HTBC or LTBC) based on a bone marrow aspirate or biopsy before infusion. HTBC patients received a single dose of tocilizumab (8-12 mg/kg) after development of high, persistent fevers. LTBC patients received standard CRS management. The primary end point was the frequency of grade 4 CRS (Penn scale), with an observed rate of ≤ 5 of 15 patients in the HTBC pre-defined as clinically meaningful. In post hoc analyses, the HTBC was compared with a historical cohort of high-tumor burden patients from the initial phase I CTL019 trial. RESULTS The primary end point was met. Seventy patients were infused with CTL019, 15 in the HTBC and 55 in the LTBC. All HTBC patients received the PT intervention. The incidence of grade 4 CRS was 27% (95% CI, 8 to 55) in the HTBC and 3.6% (95% CI, 0.4 to 13) in the LTBC. The best overall response rate was 87% in the HTBC and 100% in the LTBC. Initial CTL019 expansion was greater in the HTBC than the LTBC (P < .001), but persistence was not different (P = .73). Event-free and overall survival were worse in the HTBC (P = .004, P < .001, respectively). In the post hoc analysis, grade 4 CRS was observed in 27% versus 50% of patients in the PT and prior phase I cohorts, respectively (P = .18). CONCLUSION Risk-adapted PT administration resulted in a decrease in the expected incidence of grade 4 CRS, meeting the study end point, without adversely impacting the antitumor efficacy or safety of CTL019.
Collapse
Affiliation(s)
- Stephan Kadauke
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Regina M Myers
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Yimei Li
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Richard Aplenc
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Diane Baniewicz
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA
| | - David M Barrett
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Allison Barz Leahy
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Colleen Callahan
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Joseph G Dolan
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Whitney Gladney
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Simon F Lacey
- Abramson Cancer Center and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA
| | - Hongyan Liu
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Shannon L Maude
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Regina McGuire
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Laura S Motley
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA
| | - David T Teachey
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gerald B Wertheim
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Lisa Wray
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amanda M DiNofia
- Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephan A Grupp
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Cellular Therapy and Transplant Section and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
46
|
Farren TW, Sadanand KS, Agrawal SG. Highly Sensitive and Accurate Assessment of Minimal Residual Disease in Chronic Lymphocytic Leukemia Using the Novel CD160-ROR1 Assay. Front Oncol 2020; 10:597730. [PMID: 33344247 PMCID: PMC7744938 DOI: 10.3389/fonc.2020.597730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022] Open
Abstract
Undetectable minimal residual disease (MRD) in Chronic Lymphocytic Leukemia (CLL) has a favorable prognostic outcome compared with MRD that can be detected. This study investigated a flow cytometric assay (CD160-ROR1FCA) targeting the tumor-specific antigens CD160 and receptor tyrosine kinase-like orphan receptor 1 (ROR1), along with CD2, CD5, CD19, CD45. CD160-ROR1FCA was compared with the originally published 8-colour European Research Initiative for CLL (ERIC) gold-standard assay for CLL MRD detection. CD160-ROR1FCA had a limit of detection of 0.001% and showed strong correlation with ERIC (R = 0.98, p < 0.01) with negligible differences in MRD detection (bias -0.3152 95%CI 5.586 to -6.216). Using CD160-ROR1FCA, increased expression of both CD160 and ROR1 was found in Monoclonal B cell Lymphocytosis (MBL) compared to low-level polyclonal B-cell expansions (p < 0.01). Patients in CR and with undetectable MRD had a longer EFS (not reached) than those in CR but with detectable MRD (756 days, p < 0.01) versus 113 days in patients with partial remission (p < 0.01). Patients with MRD levels of >0.01 to 0.1% had a longer EFS (2,333 days), versus levels between 0.1 to 1% (1,049 days). CD160-ROR1FCA is a novel assay for routine CLL MRD measurement and for MBL detection. MRD status assessed by CD160-ROR1FCA after CLL treatment correlated with EFS.
Collapse
Affiliation(s)
- Timothy W Farren
- Department of Haemato-Oncology and Immunophenotyping (SIHMDS), Barts Health NHS Trust, London, United Kingdom.,Immunobiology, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Kaushik S Sadanand
- Immunobiology, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Samir G Agrawal
- Department of Haemato-Oncology and Immunophenotyping (SIHMDS), Barts Health NHS Trust, London, United Kingdom.,Immunobiology, Blizard Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
47
|
Minimal residual disease assessment in acute lymphoblastic leukemia by 4-color flow cytometry: Recommendations from the MRD Working Group of the Brazilian Society of Bone Marrow Transplantation. Hematol Transfus Cell Ther 2020; 43:332-340. [PMID: 33281111 PMCID: PMC8446261 DOI: 10.1016/j.htct.2020.09.148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/17/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022] Open
Abstract
Introduction The minimal residual disease (MRD) status plays a crucial role in the treatment of acute lymphoblastic leukemia (ALL) and is currently used in most therapeutic protocols to guide the appropriate therapeutic decision. Therefore, it is imperative that laboratories offer accurate and reliable results through well standardized technical processes by establishing rigorous operating procedures. Method Our goal is to propose a monoclonal antibody (MoAb) panel for MRD detection in ALL and provide recommendations intended for flow cytometry laboratories that work on 4-color flow cytometry platforms. Results and conclusion The document includes pre-analytical and analytical procedures, quality control assurance, technical procedures, as well as the information that needs to be included in the reports for clinicians.
Collapse
|
48
|
危 彤, 陈 晓, 张 陆, 张 傲, 竺 晓. [Clinical significance of minimal residual disease in B-lineage acute lymphoblastic leukemia pediatric patients with different fusion gene backgrounds]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:1279-1285. [PMID: 33327998 PMCID: PMC7735931 DOI: 10.7499/j.issn.1008-8830.2007097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/04/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To study the clinical significance of minimal residual disease (MRD) in B-lineage acute lymphoblastic leukemia (B-ALL) pediatric patients with different fusion gene backgrounds. METHODS A retrospective analysis was performed on the medical data of 441 B-ALL children who were treated from January 2008 to April 2015. Among the 441 children, 336 had negative fusion gene, 79 had positive ETV6-RUNX1 fusion gene, and 26 had positive E2A-PBX1 fusion gene. Flow cytometry was used to detect MRD, and the influence of MRD on day 15 (TP1), day 33 (TP2), and week 12 (TP3) of induction therapy on prognosis was analyzed. RESULTS In patients with negative fusion gene, the positive MRD group had significantly lower overall survival (OS) rate and event-free survival (EFS) rate (P < 0.05) and significantly higher recurrence rate and mortality rate at TP1, TP2, and TP3, compared with the negative MRD group (P < 0.05). In patients with positive ETV6-RUNX1, the positive MRD group had significantly lower OS and EFS rates (P < 0.05) and significantly higher recurrence rate and mortality rate (P < 0.05) than the negative MRD group only at TP1. In patients with positive E2A-PBX1, there were no significant differences in the OS rate, recurrence rate, and mortality rate between the positive and negative MRD groups at TP1, TP2, and TP3 (P > 0.05). CONCLUSIONS MRD has the most definite prognostic significance in pediatric B-ALL patients with negative fusion gene, while it has unsatisfactory prognostic significance in those with positive ETV6-RUNX1 or positive E2A-PBX1.
Collapse
Affiliation(s)
- 彤 危
- />中国医学科学院北京协和医学院血液病医院(中国医学科学院血液学研究所), 实验血液学国家重点实验室, 国家血液系统疾病临床医学研究中心, 天津 300020State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - 晓娟 陈
- />中国医学科学院北京协和医学院血液病医院(中国医学科学院血液学研究所), 实验血液学国家重点实验室, 国家血液系统疾病临床医学研究中心, 天津 300020State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - 陆阳 张
- />中国医学科学院北京协和医学院血液病医院(中国医学科学院血液学研究所), 实验血液学国家重点实验室, 国家血液系统疾病临床医学研究中心, 天津 300020State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - 傲利 张
- />中国医学科学院北京协和医学院血液病医院(中国医学科学院血液学研究所), 实验血液学国家重点实验室, 国家血液系统疾病临床医学研究中心, 天津 300020State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - 晓凡 竺
- />中国医学科学院北京协和医学院血液病医院(中国医学科学院血液学研究所), 实验血液学国家重点实验室, 国家血液系统疾病临床医学研究中心, 天津 300020State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
49
|
Li W, Morgan R, Nieder R, Truong S, Habeebu SSM, Ahmed AA. Normal or reactive minor cell populations in bone marrow and peripheral blood mimic minimal residual leukemia by flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:590-601. [PMID: 33197125 DOI: 10.1002/cyto.b.21968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/17/2020] [Accepted: 10/26/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Measurable residual disease (MRD) is a strong independent poor prognostic factor for acute leukemia. Multiparameter flow cytometry (FCM) is a commonly used MRD detection method. However, FCM MRD detection is not well standardized, and the interpretation is subjective. There are normal/reactive minor cell populations in bone marrow (BM) and peripheral blood (PB), which could be confused with MRD. METHODS The FCM data of 231 BM and 44 PB pediatric samples performed in a recent 15-month period were retrospectively reviewed. These samples were from 56 B-lymphoblastic leukemia (B-ALL) patients, 11 T-lymphoblastic leukemia (T-ALL) patients, 28 acute myeloid leukemia (AML)/myelodysplastic syndrome (MDS) patients, 44 cytopenia/leukocytosis patients, and five patients with mycosis fungoides. RESULTS There were over 10 normal or reactive minor cell populations identified with certain phenotypes mimicking MRD of acute leukemia. These mimickers included CD19+ NK cells, CD22+ basophils, CD22+ dendritic cells (DCs), and plasma cells for B-ALL MRD; CD4/8 double-negative T cells, CD4/8 double-positive T cells, cytoplasmic CD3+ NK cells, CD2- T cells, CD7- T cells, CD5- gamma delta T cells, CD56+ NKT cells for T-ALL MRD; CD33+ NK cells, CD117+ NK cells, basophils, plasmacytoid DCs, non-classical monocytes, CD56+ and/or CD61+ monocytes for AML MRD. CONCLUSIONS These data confirm the presence of a variety of normal/reactive minor cell populations that could mimic MRD of acute leukemia by FCM. Recognizing these MRD mimickers is important for correct FCM MRD interpretation.
Collapse
Affiliation(s)
- Weijie Li
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Ruth Morgan
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Roxanne Nieder
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Sa Truong
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Sahibu Sultan M Habeebu
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Atif A Ahmed
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri, USA
| |
Collapse
|
50
|
Kim M, Park CJ. Minimal Residual Disease Detection in Pediatric Acute Lymphoblastic Leukemia. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2020. [DOI: 10.15264/cpho.2020.27.2.87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Miyoung Kim
- Department of Laboratory Medicine, Hallym University Sacred Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Anyang, Korea
| | - Chan-Jeoung Park
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| |
Collapse
|