1
|
Witek MA, Larkey NE, Bartakova A, Hupert ML, Mog S, Cronin JK, Vun J, August KJ, Soper SA. Microfluidic Affinity Selection of B-Lineage Cells from Peripheral Blood for Minimal Residual Disease Monitoring in Pediatric B-Type Acute Lymphoblastic Leukemia Patients. Int J Mol Sci 2024; 25:10619. [PMID: 39408948 PMCID: PMC11477226 DOI: 10.3390/ijms251910619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Assessment of minimal residual disease (MRD) is the most powerful predictor of outcome in B-type acute lymphoblastic leukemia (B-ALL). MRD, defined as the presence of leukemic cells in the blood or bone marrow, is used for the evaluation of therapy efficacy. We report on a microfluidic-based MRD (MF-MRD) assay that allows for frequent evaluation of blood for the presence of circulating leukemia cells (CLCs). The microfluidic chip affinity selects B-lineage cells, including CLCs using anti-CD19 antibodies poised on the wall of the microfluidic chip. Affinity-selected cells are released from the capture surface and can be subjected to immunophenotyping to enumerate the CLCs, perform fluorescence in situ hybridization (FISH), and/or molecular analysis of the CLCs' mRNA/gDNA. During longitudinal testing of 20 patients throughout induction and consolidation therapy, the MF-MRD performed 116 tests, while only 41 were completed with multiparameter flow cytometry (MFC-MRD) using a bone marrow aspirate, as standard-of-care. Overall, 57% MF-MRD tests were MRD(+) as defined by CLC numbers exceeding a threshold of 5 × 10-4%, which was determined to be the limit of quantitation. Above a threshold of 0.01%, MFC-MRD was positive in 34% of patients. The MF offered the advantage of the opportunity for efficiently processing small volumes of blood (2 mL), which is important in the care of pediatric patients, especially infants. The minimally invasive means of blood collection are of high value when treating patients whose MRD is typically tested using an invasive bone marrow biopsy. MF-MRD detection can be useful for stratification of patients into risk groups and monitoring of patient well-being after completion of treatment for early recognition of potential impending disease recurrence.
Collapse
Affiliation(s)
- Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA;
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
| | - Nicholas E. Larkey
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Alena Bartakova
- Biofluidica Inc., San Diego, CA 92121, USA; (A.B.); (M.L.H.)
| | | | - Shalee Mog
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
| | - Jami K. Cronin
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Judy Vun
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Keith J. August
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA;
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
- Biofluidica Inc., San Diego, CA 92121, USA; (A.B.); (M.L.H.)
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
2
|
Chea M, Rigolot L, Canali A, Vergez F. Minimal Residual Disease in Acute Myeloid Leukemia: Old and New Concepts. Int J Mol Sci 2024; 25:2150. [PMID: 38396825 PMCID: PMC10889505 DOI: 10.3390/ijms25042150] [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: 12/31/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Minimal residual disease (MRD) is of major importance in onco-hematology, particularly in acute myeloid leukemia (AML). MRD measures the amount of leukemia cells remaining in a patient after treatment, and is an essential tool for disease monitoring, relapse prognosis, and guiding treatment decisions. Patients with a negative MRD tend to have superior disease-free and overall survival rates. Considerable effort has been made to standardize MRD practices. A variety of techniques, including flow cytometry and molecular methods, are used to assess MRD, each with distinct strengths and weaknesses. MRD is recognized not only as a predictive biomarker, but also as a prognostic tool and marker of treatment efficacy. Expected advances in MRD assessment encompass molecular techniques such as NGS and digital PCR, as well as optimization strategies such as unsupervised flow cytometry analysis and leukemic stem cell monitoring. At present, there is no perfect method for measuring MRD, and significant advances are expected in the future to fully integrate MRD assessment into the management of AML patients.
Collapse
Affiliation(s)
- Mathias Chea
- Laboratoire d’Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (M.C.); (L.R.); (A.C.)
| | - Lucie Rigolot
- Laboratoire d’Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (M.C.); (L.R.); (A.C.)
- School of Medicine, Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Alban Canali
- Laboratoire d’Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (M.C.); (L.R.); (A.C.)
- School of Medicine, Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Francois Vergez
- Laboratoire d’Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (M.C.); (L.R.); (A.C.)
- School of Medicine, Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| |
Collapse
|
3
|
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
|
4
|
Kovach AE, Wood BL. Updates on lymphoblastic leukemia/lymphoma classification and minimal/measurable residual disease analysis. Semin Diagn Pathol 2023; 40:457-471. [PMID: 37953192 DOI: 10.1053/j.semdp.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023]
Abstract
Lymphoblastic leukemia/lymphoma (ALL/LBL), especially certain subtypes, continues to confer morbidity and mortality despite significant therapeutic advances. The pathologic classification of ALL/LBL, especially that of B-ALL, has recently substantially expanded with the identification of several distinct and prognostically important genetic drivers. These discoveries are reflected in both current classification systems, the World Health Organization (WHO) 5th edition and the new International Consensus Classification (ICC). In this article, novel subtypes of B-ALL are reviewed, including DUX4, MEF2D and ZNF384-rearranged B-ALL; the rare pediatric entity B-ALL with TLF3::HLF, now added to the classifications, is discussed; updates to the category of B-ALL with BCR::ABL1-like features (Ph-like B-ALL) are summarized; and emerging genetic subtypes of T-ALL are presented. The second half of the article details current approaches to minimal/measurable residual disease (MRD) detection in B-ALL and T-ALL and presents anticipated challenges to current approaches in the burgeoning era of antigen-directed immunotherapy.
Collapse
Affiliation(s)
- Alexandra E Kovach
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, United States; Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.
| | - Brent L Wood
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, United States; Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
5
|
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
|
6
|
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: 0] [Impact Index Per Article: 0] [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
|
7
|
Robinson TM, Bowman RL, Persaud S, Liu Y, Neigenfind R, Gao Q, Zhang J, Sun X, Miles LA, Cai SF, Sciambi A, Llanso A, Famulare C, Goldberg A, Dogan A, Roshal M, Levine RL, Xiao W. Single-cell genotypic and phenotypic analysis of measurable residual disease in acute myeloid leukemia. SCIENCE ADVANCES 2023; 9:eadg0488. [PMID: 37729414 PMCID: PMC10881057 DOI: 10.1126/sciadv.adg0488] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 07/21/2023] [Indexed: 09/22/2023]
Abstract
Measurable residual disease (MRD), defined as the population of cancer cells that persist following therapy, serves as the critical reservoir for disease relapse in acute myeloid leukemia and other malignancies. Understanding the biology enabling MRD clones to resist therapy is necessary to guide the development of more effective curative treatments. Discriminating between residual leukemic clones, preleukemic clones, and normal precursors remains a challenge with current MRD tools. Here, we developed a single-cell MRD (scMRD) assay by combining flow cytometric enrichment of the targeted precursor/blast population with integrated single-cell DNA sequencing and immunophenotyping. Our scMRD assay shows high sensitivity of approximately 0.01%, deconvolutes clonal architecture, and provides clone-specific immunophenotypic data. In summary, our scMRD assay enhances MRD detection and simultaneously illuminates the clonal architecture of clonal hematopoiesis/preleukemic and leukemic cells surviving acute myeloid leukemia therapy.
Collapse
Affiliation(s)
- Troy M. Robinson
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert L. Bowman
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sonali Persaud
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying Liu
- Department of Pathology and Laboratory Medicine, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Molecular Diagnostic Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rosemary Neigenfind
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qi Gao
- Department of Pathology and Laboratory Medicine, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jingping Zhang
- Department of Pathology and Laboratory Medicine, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiaotian Sun
- Department of Pathology and Laboratory Medicine, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Linde A. Miles
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sheng F. Cai
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Christopher Famulare
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aaron Goldberg
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mikhail Roshal
- Department of Pathology and Laboratory Medicine, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ross L. Levine
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wenbin Xiao
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
8
|
Lucas F, Hergott CB. Advances in Acute Myeloid Leukemia Classification, Prognostication and Monitoring by Flow Cytometry. Clin Lab Med 2023; 43:377-398. [PMID: 37481318 DOI: 10.1016/j.cll.2023.04.005] [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: 07/24/2023]
Abstract
Although final classification of acute myeloid leukemia (AML) integrates morphologic, cytogenetic, and molecular data, flow cytometry remains an essential component of modern AML diagnostics. Here, we review the current role of flow cytometry in the classification, prognostication, and monitoring of AML. We cover immunophenotypic features of key genetically defined AML subtypes and their effects on biological and clinical behaviors, review clinically tractable strategies to differentiate leukemias with ambiguous immunophenotypes more accurately and discuss key principles of standardization for measurable residual disease monitoring. These advances underscore flow cytometry's continued growth as a powerful diagnostic, management, and discovery tool.
Collapse
Affiliation(s)
- Fabienne Lucas
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Christopher B Hergott
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Zhang J, Oak J. Challenges of detecting measurable/minimal disease in acute leukemia. Semin Diagn Pathol 2023; 40:216-220. [PMID: 37150656 DOI: 10.1053/j.semdp.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023]
Abstract
Measurable/minimal residual disease (MRD) tracking has emerged as a powerful tool for assessing treatment response and predicting outcomes in acute leukemia. However, the clinical and technological challenges associated with MRD tracking must be addressed to improve its utility in routine patient care. This review article aims to provide a summary of the different MRD methodologies used in acute leukemia. It highlights the strengths, diagnostic pitfalls, and clinical utility associated with MRD tracking in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Pathology, 300 Pasteur Drive, L235, Stanford, CA 94305, United States
| | - Jean Oak
- Department of Pathology, 300 Pasteur Drive, L235, Stanford, CA 94305, United States.
| |
Collapse
|
10
|
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
|
11
|
Semchenkova A, Zhogov V, Zakharova E, Mikhailova E, Illarionova O, Larin S, Novichkova G, Karachunskiy A, Maschan M, Popov A. Flow cell sorting followed by PCR-based clonality testing may assist in questionable diagnosis and monitoring of acute lymphoblastic leukemia. Int J Lab Hematol 2023. [PMID: 36871952 DOI: 10.1111/ijlh.14053] [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: 07/26/2022] [Accepted: 02/21/2023] [Indexed: 03/07/2023]
Abstract
INTRODUCTION Multicolor flow cytometry (MFC) has highly reliable and flexible algorithms for diagnosis and monitoring of acute lymphoblastic leukemia (ALL). However, MFC analysis can be affected by poor sample quality or novel therapeutic options (e.g., targeted therapies and immunotherapy). Therefore, an additional confirmation of MFC data may be needed. We propose a simple approach for validation of MFC findings in ALL by sorting questionable cells and analyzing immunoglobulin/T-cell receptor (IG/TR) gene rearrangements via EuroClonality-based multiplex PCR. PATIENTS AND METHODS We obtained questionable MFC results for 38 biological samples from 37 patients. In total, 42 cell populations were isolated by flow cell sorting for downstream multiplex PCR. Most of the patients (n = 29) had B-cell precursor ALL and were investigated for measurable residual disease (MRD); 79% of them received CD19-directed therapy (blinatumomab or CAR-T). RESULTS We established the clonal nature of 40 cell populations (95.2%). By using this technique, we confirmed very low MRD levels (<0.01% MFC-MRD). We also applied it to several ambiguous findings for diagnostic samples, including those with mixed-phenotype acute leukemia, and the results obtained impacted the final diagnosis. CONCLUSION We have demonstrated possibilities of a combined approach (cell sorting and PCR-based clonality assessment) to validate MFC findings in ALL. The technique is easy to implement in diagnostic and monitoring workflows, as it does not require isolation of a large number of cells and knowledge of individual clonal rearrangements. We believe it provides important information for further treatment.
Collapse
Affiliation(s)
- Alexandra Semchenkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Vladimir Zhogov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Elena Zakharova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Ekaterina Mikhailova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Olga Illarionova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Sergey Larin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Galina Novichkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexander Karachunskiy
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Michael Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexander Popov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
12
|
Kovach AE, Raca G. Modern Classification and Management of Pediatric B-cell Leukemia and Lymphoma. Surg Pathol Clin 2023; 16:249-266. [PMID: 37149359 DOI: 10.1016/j.path.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Although pediatric hematopathology overlaps with that of adults, certain forms of leukemia and lymphoma, and many types of reactive conditions affecting the bone marrow and lymph nodes, are unique to children. As part of this series focused on lymphomas, this article (1) details the novel subtypes of lymphoblastic leukemia seen primarily in children and described since the 2017 World Health Organization classification and (2) discusses unique concepts in pediatric hematopathology, including nomenclature changes and evaluation of surgical margins in selected lymphomas.
Collapse
Affiliation(s)
- Alexandra E Kovach
- Division of Laboratory Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA; Clinical Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA.
| | - Gordana Raca
- Clinical Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA; Division of Genomic Medicine, Department of Pathology and Laboratory Medicine, Center for Personalized Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
13
|
Choi JK, Mead PE. Laboratory Aspects of Minimal / Measurable Residual Disease Testing in B-Lymphoblastic Leukemia. Clin Lab Med 2023; 43:115-125. [PMID: 36764804 DOI: 10.1016/j.cll.2022.09.022] [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: 02/11/2023]
Abstract
Minimal residual disease detection provides critical prognostic predictor of treatment outcome and is the standard of care for B lymphoblastic leukemia. Flow cytometry-based minimal residual disease detection is the most common test modality and has high sensitivity (0.01%) and a rapid turnaround time (24 hours). This article details the leukemia associated immunophenotype analysis approach for flow cytometry-based minimal residual disease detection used at St. Jude Children's Research Hospital and importance of using guide gates and back-gating.
Collapse
Affiliation(s)
- John Kim Choi
- Division of Laboratory Medicine, The University of Alabama at Birmingham, WP P230N, 619 19th Street South, Birmingham, AL 35249-7331, USA.
| | - Paul E Mead
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, D4026G, Mailstop 342, Memphis, TN 38105, USA
| |
Collapse
|
14
|
Abd El Monem M, El Ashry R, Bassiouny MR, Aref S, Abd El Mabood S. The prognostic significance of cytokine receptor-like factor 2 expression and <i>JAK2</i> mutation in pediatric B-cell acute lymphoblastic leukemia: A prospective cohort study. PEDIATRIC HEMATOLOGY/ONCOLOGY AND IMMUNOPATHOLOGY 2023; 22:40-45. [DOI: 10.24287/1726-1708-2023-22-1-40-45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy. Philadelphia (Ph)-like B-cell acute lymphoblastic leukemia (B-ALL) is defined by a gene expression profile similar to Phpositive B-ALL and shows a large number of genetic alterations in the cytokine receptor and kinasesignaling pathway genes that contribute to its aggressive phenotype and frequent disease recurrence – the main cause of death in affected children. Here, we aimed to correlate CRLF2 expression and JAK2 mutations in B-ALL patients with other prognostic factors and the patients’ outcomes as well as to evaluate their prognostic significance. The study was approved by the local institutional review board and written consents were obtained from a parent of each child before their enrolment. We included 54 newly diagnosed B-ALL pediatric patients (median age: 9.0 (2.0–18.0)) who were stratified either into a standard-risk (SR) or high-risk (HR) group and treated according to the modified BerlinFrankfurt-Münster 90 protocol (ALL-BFM 90). Fresh bone marrow samples were used to determine CRLF2 expression as well as to search for the JAK2 V617F mutation. Normal CRLF2 expression was reported in the SR patients much more often than in the HR group, while its overexpression was more common in the HR patients than in the SR ones (22 vs 6 and 18 vs 8, respectively, p < 0.001). CRLF2 was also more often overexpressed in the MRD-positive cases than in the negative ones (17 vs 9, p < 0.001), while normal CRLF2 expression was more common in the MRD-negative patients compared to the MRD-positive ones (24 vs 4, p < 0.001) which supports the unfavorable prognostic value of CRLF2 in relation to MRD positivity at the end of the induction treatment. JAK2 mutation was detected only in 2 patients belonging to the CRLF2 overexpression group which made the assessment of the prognostic significance of this mutation impossible. Notably, none of the patients with normal CRLF2 expression ended up relapsing while 4 patients with overexpressed CRLF2 developed a relapse (p = 0.031). The study subjects were followed up for up to 24 months, and we did not find CRLF2 overexpression to negatively influence overall survival, however, it did have an adverse effect on relapse-free survival. In summary, CRLF2 overexpression was found to be an unfavorable prognostic factor in childhood ALL as it was expressed more in high-risk patients and in those with poor treatment response. The analysis of CRLF2 expression in B-ALL pediatric patients may help in risk stratification and can potentially offer new treatment options based on novel CRLF2 inhibitors.
Collapse
|
15
|
Chen YF, Li J, Xu LL, Găman MA, Zou ZY. Allogeneic stem cell transplantation in the treatment of acute myeloid leukemia: An overview of obstacles and opportunities. World J Clin Cases 2023; 11:268-291. [PMID: 36686358 PMCID: PMC9850970 DOI: 10.12998/wjcc.v11.i2.268] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/02/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
As an important treatment for acute myeloid leukemia, allogeneic hematopoietic stem cell transplantation (allo-HSCT) plays an important role in reducing relapse and improving long-term survival. With rapid advancements in basic research in molecular biology and immunology and with deepening understanding of the biological characteristics of hematopoietic stem cells, allo-HSCT has been widely applied in clinical practice. During allo-HSCT, preconditioning, the donor, and the source of stem cells can be tailored to the patient’s conditions, greatly broadening the indications for HSCT, with clear survival benefits. However, the risks associated with allo-HSCT remain high, i.e. hematopoietic reconstitution failure, delayed immune reconstitution, graft-versus-host disease, and post-transplant relapse, which are bottlenecks for further improvements in allo-HSCT efficacy and have become hot topics in the field of HSCT. Other bottlenecks recognized in the current treatment of individuals diagnosed with acute myeloid leukemia and subjected to allo-HSCT include the selection of the most appropriate conditioning regimen and post-transplantation management. In this paper, we reviewed the progress of relevant research regarding these aspects.
Collapse
Affiliation(s)
- Yong-Feng Chen
- Department of Basic Medical Sciences, School of Medicine of Taizhou University, Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Jing Li
- Department of Histology and Embryology, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Ling-Long Xu
- Department of Hematology, Taizhou Central Hospital, Taizhou 318000, Zhejiang Province, China
| | - Mihnea-Alexandru Găman
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Zhen-You Zou
- Department of Scientific Research,Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
16
|
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
|
17
|
Ganzel C, Sun Z, Baslan T, Zhang Y, Gönen M, Abdel-Wahab OI, Racevskis J, Garrett-Bakelman F, Lowe SW, Fernandez HF, Ketterling R, Luger SM, Litzow M, Lazarus HM, Rowe JM, Tallman MS, Levine RL, Paietta E. Measurable residual disease by flow cytometry in acute myeloid leukemia is prognostic, independent of genomic profiling. Leuk Res 2022; 123:106971. [PMID: 36332294 PMCID: PMC9789386 DOI: 10.1016/j.leukres.2022.106971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/04/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
Abstract
Measurable residual disease (MRD) assessment provides a potent indicator of the efficacy of anti-leukemic therapy. It is unknown, however, whether integrating MRD with molecular profiling better identifies patients at risk of relapse. To investigate the clinical relevance of MRD in relation to a molecular-based prognostic schema, we measured MRD by flow cytometry in 189 AML patients enrolled in ECOG-ACRIN E1900 trial (NCT00049517) in morphologic complete remission (CR) (28.8 % of the original cohort) representing 44.4 % of CR patients. MRD positivity was defined as ≥ 0.1 % of leukemic bone marrow cells. Risk classification was based on standard cytogenetics, fluorescence-in-situ-hybridization, somatic gene analysis, and sparse whole genome sequencing for copy number ascertainment. At 84.6 months median follow-up of patients still alive at the time of analysis (range 47.0-120 months), multivariate analysis demonstrated that MRD status at CR (p = 0.001) and integrated molecular risk (p = 0.0004) independently predicted overall survival (OS). Among risk classes, MRD status significantly affected OS only in the favorable risk group (p = 0.002). Expression of CD25 (α-chain of the interleukin-2 receptor) by leukemic myeloblasts at diagnosis negatively affected OS independent of post-treatment MRD levels. These data suggest that integrating MRD with genetic profiling and pre-treatment CD25 expression may improve prognostication in AML.
Collapse
Affiliation(s)
- Chezi Ganzel
- Hematology Department, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| | - Zhuoxin Sun
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar I Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Janis Racevskis
- Department of Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Francine Garrett-Bakelman
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Departments of Medicine and Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hugo F Fernandez
- Malignant Hematology and Cellular Therapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Rhett Ketterling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Selina M Luger
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Litzow
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jacob M Rowe
- Hematology Department, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Martin S Tallman
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
18
|
Mikhailova E, Illarionova O, Komkov A, Zerkalenkova E, Mamedov I, Shelikhova L, Olshanskaya Y, Miakova N, Novichkova G, Karachunskiy A, Maschan M, Popov A. Reliable Flow-Cytometric Approach for Minimal Residual Disease Monitoring in Patients with B-Cell Precursor Acute Lymphoblastic Leukemia after CD19-Targeted Therapy. Cancers (Basel) 2022; 14:5445. [PMID: 36358863 PMCID: PMC9658935 DOI: 10.3390/cancers14215445] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 09/10/2023] Open
Abstract
We aimed to develop an antibody panel and data analysis algorithm for multicolor flow cytometry (MFC), which is a reliable method for minimal residual disease (MRD) detection in patients with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) treated with CD19-directed therapy. The development of the approach, which was adapted for the case of possible CD19 loss, was based on the additional B-lineage marker expression data obtained from a study of primary BCP-ALL patients, an analysis of the immunophenotypic changes that occur during blinatumomab or CAR-T therapy, and an analysis of very early CD19-negative normal BCPs. We have developed a single-tube 11-color panel for MFC-MRD detection. CD22- and iCD79a-based primary B-lineage gating (preferably consecutive) was recommended. Based on patterns of antigen expression changes and the relative expansion of normal CD19-negative BCPs, guidelines for MFC data analysis and interpretation were established. The suggested approach was tested in comparison with the molecular techniques: IG/TR gene rearrangement detection by next-generation sequencing (NGS) and RQ-PCR for fusion-gene transcripts (FGTs). Qualitative concordance rates of 82.8% and 89.8% were obtained for NGS-MRD and FGT-MRD results, respectively. We have developed a sensitive and reliable approach that allows MFC-MRD monitoring after CD19-directed treatment, even in the case of possible CD19 loss.
Collapse
Affiliation(s)
- Ekaterina Mikhailova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Olga Illarionova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Alexander Komkov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117998 Moscow, Russia
| | - Elena Zerkalenkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Ilgar Mamedov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117998 Moscow, Russia
| | - Larisa Shelikhova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Yulia Olshanskaya
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Natalia Miakova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Galina Novichkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Alexander Karachunskiy
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Michael Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| | - Alexander Popov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117998 Moscow, Russia
| |
Collapse
|
19
|
Fernando F, Robertson HF, El-Zahab S, Pavlů J. How I Use Measurable Residual Disease in the Clinical Management of Adult Acute Lymphoblastic Leukemia. Clin Hematol Int 2021; 3:130-141. [PMID: 34938985 PMCID: PMC8690704 DOI: 10.2991/chi.k.211119.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/05/2021] [Indexed: 11/01/2022] Open
Abstract
Over the last decade the use of measurable residual disease (MRD) diagnostics in adult acute lymphoblastic leukemia (ALL) has expanded from a limited number of study groups in Europe and the United States to a world-wide application. In this review, we summarize the advantages and drawbacks of the current available techniques used for MRD monitoring. Through the use of three representative case studies, we highlight the advances in the use of MRD in clinical decision-making in the management of ALL in adults. We acknowledge discrepancies in MRD monitoring and treatment between different countries, reflecting differing availability, accessibility and affordability.
Collapse
Affiliation(s)
- Fiona Fernando
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | | | - Sarah El-Zahab
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Jiří Pavlů
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| |
Collapse
|
20
|
Ayyanar P, Kar R, Dubashi B, Basu D. Post-chemotherapy Changes in Bone Marrow in Acute Leukemia With Emphasis on Detection of Residual Disease by Immunohistochemistry. Cureus 2021; 13:e20175. [PMID: 35004001 PMCID: PMC8725173 DOI: 10.7759/cureus.20175] [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] [Accepted: 12/05/2021] [Indexed: 11/17/2022] Open
Abstract
Introduction In acute leukemia, the leading cause of treatment failure is disease relapse leading to a low level of complete remission and short overall survival. Post-chemotherapy marrow examination gives vital clues regarding treatment response and marrow regeneration. Aim We aimed to study the histomorphological changes in post-chemotherapy bone marrow in acute leukemias, monitor residual disease by immunohistochemistry (IHC) on trephine biopsy, and correlate survival status. Method This study was a prospective clinical study. A total of 155 post-induction cases (acute myeloid leukemia [AML] - 68 and acute lymphoblastic leukemia [ALL] - 87), from January 2014 to December 2015, were included with a follow-up of 4-28 months. A detailed histomorphology was studied in all cases. IHC was applied in 88 cases of post-induction marrow, which showed morphologic suspicion of an increase in blasts. Observations Post-induction marrow was hypercellular in 55.9% of AML and normocellular in 56.3% of ALL. Regenerative hematopoiesis was noted in 37.4% of AML and 88.5% of cases of ALL. Marrow serous atrophy and stromal edema were associated with delayed recovery of counts and their recovery duration ranged from one to five months. Twenty-seven bone marrow aspirates were unsatisfactory, and their trephine biopsies were showed remission in 20 cases and stromal changes in nine cases. In addition, trephine biopsy picked up residual leukemic blasts in four cases in which aspirate showed remission status. Post-induction marrow IHC with scattered positivity for blasts showed sustained remission in 96% cases, and in those with clustered positivity, 28.6% showed residual disease, and 7.2% showed relapse at the end of the study period. The median survival duration was 13, 3, and 12 months for cases with sustained remission, residual disease, and relapse, respectively. There was a statistically significant difference in median survival of patients in the three groups (sustained remission, residual disease, and relapse) (p=0.000). Conclusion We conclude that histomorphology augmented by IHC on trephine biopsy gives valuable information regarding post-chemotherapy changes and residual disease status. Bone marrow trephine biopsy is an important tool to assess the remission status of patients with acute leukemia.
Collapse
|
21
|
Rosenberg CA, Bill M, Maguire O, Petersen MA, Kjeldsen E, Hokland P, Ludvigsen M. Imaging flow cytometry reveals a subset of TdT negative T-ALL blasts with very low forward scatter on conventional flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2021; 102:107-114. [PMID: 34648681 DOI: 10.1002/cyto.b.22035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Studies in T-cell acute lymphoblastic leukemia (T-ALL) have shown that leukemic blast populations may display immunophenotypic heterogeneity. In the clinical setting, evaluation of measurable residual disease during treatment and follow-up is highly dependent on knowledge of the diversity of blast subsets. Here, we set out to evaluate whether variation in expression of the blast marker, TdT, in T-ALL blasts could correspond to differences in morphometric features. METHODS We investigated diagnostic bone marrow samples from six individual T-ALL patients run in parallel on imaging flow cytometry (IFC) and conventional flow cytometry (CFC). RESULTS Guided by the imagery available in IFC, we identified distinct TdTneg and TdTpos subpopulations with apparent differences in internal complexity. As TdTneg blasts predominantly displayed very low forward scatter (FSC) on CFC, these subsets were initially excluded from routine analysis as debris, elements of small diameter, apoptotic, and/or dead cells. However, IFC-based morphometric analyses demonstrated that cell size and shape of TdTneg blasts were comparable to the TdTpos cells and without morphometric apoptotic hallmarks, supporting that the TdTneg subpopulation corresponded to T-ALL blasts. Fluorescence in situ hybridization analyses substantiated the clinical relevance of TdTneg FSCvery-low cells by retrieving known diagnostic cytogenetic abnormalities at comparable frequencies in purified TdTneg FSCvery-low and TdTpos FSCint subsets. CONCLUSION We highlight this finding as knowledge of phenotypic heterogeneity is of crucial importance in the clinical setting for delineation and quantification of blast subpopulations of potential biological relevance. We argue that the IFC imagery may allow for visual verification and improvement of applied gating strategies.
Collapse
Affiliation(s)
| | - Marie Bill
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Orla Maguire
- Flow and Image Cytometry Shared Resource, Roswell Park Cancer Comprehensive Cancer Center, Buffalo, New York, USA
| | - Marianne A Petersen
- Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Eigil Kjeldsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Hokland
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Katz SG, Edappallath S, Xu ML. IRF8 is a Reliable Monoblast Marker for Acute Monocytic Leukemias. Am J Surg Pathol 2021; 45:1391-1398. [PMID: 34172624 DOI: 10.1097/pas.0000000000001765] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Blast evaluation in patients with acute monocytic leukemias (AMoL) is notoriously difficult due to the lack of reliable surface markers and cytologic subtleties on the aspirate smears. While blasts of most nonmonocytic acute leukemias express CD34, available immunohistochemical antibodies to monocytic blasts also mark normal background mature monocytes. We searched for a potential biomarker candidate by surveying specific gene expression profiles of monocyte progenitors. Our investigations led us to IRF8, which is a lineage-specific transcription factor critical for the production of monocytic and dendritic cell progenitors. In this study, we tested and validated a monoclonal antibody to IRF8 as a novel immunohistochemical stain for trephine core biopsies of human bone marrow. We assessed the expression of IRF8 in 90 cases of AMoL, including posttherapy staging bone marrows, 23 cases of chronic myelomonocytic leukemia, 26 cases of other acute myeloid leukemia subtypes, and 18 normal control marrows. In AMoL, there was high correlation of IRF8-positive cells to aspirate blast count (R=0.95). Comparison of IRF8 staining to aspirate blast percentage in chronic myelomonocytic leukemia also showed good correlation (R=0.86). In contrast, IRF8-positive cells did not correlate with blast count in other subtypes of acute myeloid leukemia (R=0.56) and staining was <5% in all normal control marrows, even those with reactive monocytosis. We found that IRF8 was also weakly reactive in B cells and hematogones, with the latter accounting for rare cases of discrepancies. When IRF8 was used to categorize cases as AMoL, positive for residual leukemia or negative, the sensitivity was 98%, specificity was 82%, positive predictive value was 86%, and negative predictive value was 98%. These results demonstrate that IRF8 may serve as a clinically useful immunostain to diagnose and track AMoLs on bone marrow core biopsies. This can be particularly impactful in the setting of poor aspiration and focal blast increase. In the era of new targeted therapies that have been reported to induce monocytic outgrowths of leukemia, a marker for malignant monoblasts may prove even more critical.
Collapse
Affiliation(s)
- Samuel G Katz
- Department of Pathology, Yale New-Haven Hospital, Yale School of Medicine, New Haven, CT
| | | | | |
Collapse
|
23
|
F McGowan P, D Hyter S, Cui W, Plummer RM, Godwin AK, Zhang D. Comparison of flow cytometry and next-generation sequencing in minimal residual disease monitoring of acute myeloid leukemia: One institute's practical clinical experience. Int J Lab Hematol 2021; 44:118-126. [PMID: 34585519 DOI: 10.1111/ijlh.13711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/19/2021] [Accepted: 09/09/2021] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Monitoring patients with acute myeloid leukemia can be implemented through various techniques such as multiparameter flow cytometry, real-time quantitative polymerase chain reaction, and next-generation sequencing. However, there is scarce studies when comparing the data of next-generation sequencing and flow cytometry for monitoring disease progression, particularly how they might supplement one another when used in tandem. METHODS We investigated 107 patients via retrospective analysis using follow-up MFC and NGS data with a total of 717 MFC and 247 NGS studies to compare these methods in monitoring minimal/measurable residual disease. RESULTS 197 instances were MFC+ /NGS+ , 3 were MFC- /NGS- , 44 were MFC- /NGS+ , and 3 are MFC+ /NGS- . The majority of the MFC- /NGS+ cases occurred within 6 months during the post-treatment phase (64%). Among 44 MFC- /NGS+ instances, 13 had similar NGS profiles to their original day 0 diagnosis. The remaining cases showed preleukemic clonal hematopoiesis mutations, "likely pathogenic mutations," or "variants of uncertain significance." CONCLUSION Our findings show that flow cytometry has its advantages with comparable sensitivity in detecting minimal/measurable residual disease. Next-generation sequencing could be used in an increased and more regular capacity in conjunction with flow cytometry to achieve a more comprehensive surveillance of these patients, resulting in improved outcomes.
Collapse
Affiliation(s)
- Paul F McGowan
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Stephen D Hyter
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wei Cui
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Regina M Plummer
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Andrew K Godwin
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Da Zhang
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
24
|
Das N, Gupta R, Gupta SK, Bakhshi S, Seth R, Kumar C, Rai S, Singh S, Prajapati VK, Gogia A, Sahoo RK, Sharma A, Kumar L. Critical evaluation of the utility of pre- and post-therapy immunophenotypes in assessment of measurable residual disease in B-ALL. Ann Hematol 2021; 100:2487-2500. [PMID: 34236495 DOI: 10.1007/s00277-021-04580-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 06/15/2021] [Indexed: 10/20/2022]
Abstract
Measurable residual disease (MRD) is an important parameter to predict outcome in B-cell acute lymphoblastic leukemia (B-ALL). Two different approaches have been used for the assessment of MRD by multiparametric flow cytometry that include the "Leukemia Associated Aberrant Immunophenotype (LAIP)" and "Difference from Normal (DFN)" approach. In this retrospective study, we analyzed 539 samples obtained from 281 patients of which 258 were paired samples and the remaining 23 samples were from post-induction time point only, to explore the utility of baseline immunophenotype (IPT) for MRD assessment. Single-tube 10-color panel was used both at diagnosis and MRD time points. Out of 281 patients, 31.67% (n = 89) were positive and 68.32% (n = 192) were negative for MRD. Among 258 paired diagnostic and follow-up samples, baseline IPT was required in only 9.31% (24/258) cases which included cases with hematogone pattern and isolated dim to negative CD10 expression patterns. Comparison of baseline IPT with post-induction MRD positive samples showed a change in expression of at least one antigen in 94.04% cases. Although the immunophenotypic change in expression of various antigens is frequent in post-induction samples of B-ALL, it does not adversely impact the MRD assessment. In conclusion, the baseline IPT is required in less than 10% of B-ALL, specifically those with hematogone pattern and/or dim to negative expression of CD10. Hence, a combination of DFN and LAIP approach is recommended for reliable MRD assessment.
Collapse
Affiliation(s)
- Nupur Das
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India.
| | - Sanjeev Kumar Gupta
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| | - Rachna Seth
- Department of Pediatrics, AIIMS, New Delhi, India
| | - Chandan Kumar
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Sandeep Rai
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Saroj Singh
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Vijay Kumar Prajapati
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Ajay Gogia
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| | - Atul Sharma
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| | - Lalit Kumar
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| |
Collapse
|
25
|
Choi JK, Mead PE. Laboratory Aspects of Minimal / Measurable Residual Disease Testing in B-Lymphoblastic Leukemia. Clin Lab Med 2021; 41:485-495. [PMID: 34304777 DOI: 10.1016/j.cll.2021.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Minimal residual disease detection provides critical prognostic predictor of treatment outcome and is the standard of care for B lymphoblastic leukemia. Flow cytometry-based minimal residual disease detection is the most common test modality and has high sensitivity (0.01%) and a rapid turnaround time (24 hours). This article details the leukemia associated immunophenotype analysis approach for flow cytometry-based minimal residual disease detection used at St. Jude Children's Research Hospital and importance of using guide gates and back-gating.
Collapse
Affiliation(s)
- John Kim Choi
- Division of Laboratory Medicine, The University of Alabama at Birmingham, WP P230N, 619 19th Street South, Birmingham, AL 35249-7331, USA.
| | - Paul E Mead
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, D4026G, Mailstop 342, Memphis, TN 38105, USA
| |
Collapse
|
26
|
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
|
27
|
Hanekamp D, Tettero JM, Ossenkoppele GJ, Kelder A, Cloos J, Schuurhuis GJ. AML/Normal Progenitor Balance Instead of Total Tumor Load (MRD) Accounts for Prognostic Impact of Flowcytometric Residual Disease in AML. Cancers (Basel) 2021; 13:2597. [PMID: 34073205 PMCID: PMC8198261 DOI: 10.3390/cancers13112597] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Measurable residual disease (MRD) in AML, assessed by multicolor flow cytometry, is an important prognostic factor. Progenitors are key populations in defining MRD, and cases of MRD involving these progenitors are calculated as percentage of WBC and referred to as white blood cell MRD (WBC-MRD). Two main compartments of WBC-MRD can be defined: (1) the AML part of the total primitive/progenitor (CD34+, CD117+, CD133+) compartment (referred to as primitive marker MRD; PM-MRD) and (2) the total progenitor compartment (% of WBC, referred to as PM%), which is the main quantitative determinant of WBC-MRD. Both are related as follows: WBC-MRD = PM-MRD × PM%. We explored the relative contribution of each parameter to the prognostic impact. In the HOVON/SAKK study H102 (300 patients), based on two objectively assessed cut-off points (2.34% and 10%), PM-MRD was found to offer an independent prognostic parameter that was able to identify three patient groups with different prognoses with larger discriminative power than WBC-MRD. In line with this, the PM% parameter itself showed no prognostic impact, implying that the prognostic impact of WBC-MRD results from the PM-MRD parameter it contains. Moreover, the presence of the PM% parameter in WBC-MRD may cause WBC-MRD false positivity and WBC-MRD false negativity. For the latter, at present, it is clinically relevant that PM-MRD ≥ 10% identifies a patient sub-group with a poor prognosis that is currently classified as good prognosis MRDnegative using the European LeukemiaNet 0.1% consensus MRD cut-off value. These observations suggest that residual disease analysis using PM-MRD should be conducted.
Collapse
Affiliation(s)
- Diana Hanekamp
- Department of Hematology, Amsterdam University Medical Centers, Cancer Center VU University Medical Center, 1081 HV Amsterdam, The Netherlands; (D.H.); (J.M.T.); (G.J.O.); (A.K.); (J.C.)
- Department of Hematology, Erasmus MC, NL-3000 CA Rotterdam, The Netherlands
| | - Jesse M. Tettero
- Department of Hematology, Amsterdam University Medical Centers, Cancer Center VU University Medical Center, 1081 HV Amsterdam, The Netherlands; (D.H.); (J.M.T.); (G.J.O.); (A.K.); (J.C.)
| | - Gert J. Ossenkoppele
- Department of Hematology, Amsterdam University Medical Centers, Cancer Center VU University Medical Center, 1081 HV Amsterdam, The Netherlands; (D.H.); (J.M.T.); (G.J.O.); (A.K.); (J.C.)
| | - Angèle Kelder
- Department of Hematology, Amsterdam University Medical Centers, Cancer Center VU University Medical Center, 1081 HV Amsterdam, The Netherlands; (D.H.); (J.M.T.); (G.J.O.); (A.K.); (J.C.)
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam University Medical Centers, Cancer Center VU University Medical Center, 1081 HV Amsterdam, The Netherlands; (D.H.); (J.M.T.); (G.J.O.); (A.K.); (J.C.)
| | - Gerrit Jan Schuurhuis
- Department of Hematology, Amsterdam University Medical Centers, Cancer Center VU University Medical Center, 1081 HV Amsterdam, The Netherlands; (D.H.); (J.M.T.); (G.J.O.); (A.K.); (J.C.)
| |
Collapse
|
28
|
Zhang ZY, Huang LX, Xu ZW, Wang P, Lei Y, Liu AL. Efficient Determination of PML/RARα Fusion Gene by the Electrochemical DNA Biosensor Based on Carbon Dots/Graphene Oxide Nanocomposites. Int J Nanomedicine 2021; 16:3497-3508. [PMID: 34045854 PMCID: PMC8144022 DOI: 10.2147/ijn.s308258] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022] Open
Abstract
Purpose The PML/RARα fusion gene as a leukemogenesis plays a significant role in clinical diagnosis of the early stage of acute promyelocytic leukemia (APL). Here, we present an electrochemical biosensor for PML/RARα fusion gene detection using carbon dots functionalized graphene oxide (CDs/GO) nanocomposites modified glassy carbon electrode (CDs/GO/GCE). Materials and Methods In this work, the CDs/GO nanocomposites are produced through π-π stacking interaction and could be prepared in large quantities by a facile and economical way. The CDs/GO nanocomposites were decorated onto electrode surface to improve the electrochemical activity and as a bio-platform attracted the target deoxyribonucleic acid (DNA) probe simultaneously. Results The CDs/GO/GCE was fabricated successfully and exhibits high electrochemical activity, good biocompatibility, and strong bioaffinity toward the target DNA sequences, compared with only the pristine CDs on GCE or GO on GCE. The DNA biosensor displays excellent sensing performance for detecting the relevant pathogenic DNA of APL with a detection limit of 83 pM (S/N = 3). Conclusion According to the several experimental results, we believe that the simple and economical DNA biosensor has the potential to be an effective and powerful tool for detection of pathogenic genes in the clinical diagnosis.
Collapse
Affiliation(s)
- Zi-Yang Zhang
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Lin-Xiao Huang
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Zhi-Wei Xu
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Peng Wang
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Yun Lei
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People's Republic of China.,Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Ai-Lin Liu
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People's Republic of China.,Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| |
Collapse
|
29
|
Morphologic remission status is limited compared to ΔN flow cytometry: a Children's Oncology Group AAML0531 report. Blood Adv 2021; 4:5050-5061. [PMID: 33080007 DOI: 10.1182/bloodadvances.2020002070] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/11/2020] [Indexed: 12/18/2022] Open
Abstract
Risk stratification for acute myeloid leukemia (AML) uses molecular and cytogenetic abnormalities identified at diagnosis. Response to therapy informs risk, and morphology continues to be used more frequently than flow cytometry. Herein, the largest cohort of pediatric patients prospectively assessed for measurable residual disease (MRD) by flow cytometry (N = 784) is reported. The "difference from normal" (ΔN) technique was applied: 31% of all patients tested positive (AML range, 0.02% to 91%) after the first course of treatment on Children's Oncology Group study AAML0531. Detection of MRD following initial chemotherapy proved the strongest predicator of overall survival (OS) in univariable and multivariable analyses, and was predictive of relapse risk, disease-free survival, and treatment-related mortality. Clearance of MRD after a second round of chemotherapy did not improve survival. The morphologic definition of persistent disease (>15% AML) failed 27% of the time; those identified as MRD- had superior outcomes. Similarly, for patients not achieving morphologic remission (>5% blasts), 36% of patients were MRD- and had favorable outcomes compared with those who were MRD+ (P < .001); hence an increase in myeloid progenitor cells can be favorable when ΔN classifies them as phenotypically normal. Furthermore, ΔN reclassified 20% of patients in morphologic remission as having detectable MRD with comparable poor outcomes. Retrospective analysis using the relapse phenotype as a template demonstrated that 96% of MRD- patients had <0.02% of the relapse immunophenotype in their end of induction 1 marrow. Thus, the detection of abnormal myeloid progenitor cells by ΔN is both specific and sensitive, with a high predictive signal identifiable early in treatment. This trial was registered at www.clinicaltrials.gov as #NCT00372593.
Collapse
|
30
|
Cherian S, Soma LA. How I Diagnose Minimal/Measurable Residual Disease in B Lymphoblastic Leukemia/Lymphoma by Flow Cytometry. Am J Clin Pathol 2021; 155:38-54. [PMID: 33236071 DOI: 10.1093/ajcp/aqaa242] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Assessment for minimal/measurable residual disease (MRD) is a powerful prognostic factor in B lymphoblastic leukemia/lymphoma (B-LL/L) that is quickly becoming standard of care in assessing patients with B-LL/L posttherapy. MRD can be assessed using methodologies including flow cytometry and molecular genetics, with the former being rapid, relatively inexpensive, and widely applicable in many hematopathology/flow cytometry laboratories. METHODS This article presents an approach to MRD detection in B-LL/L by flow cytometry through case presentations with illustration of several potential pitfalls. We review normal maturation patterns, antigens used for assessment, flow panels that can be utilized, considerations to be made during therapy, and clinical impact. The benefits and drawbacks when using the "different from normal" and "leukemia associated phenotype" approaches are considered. RESULTS Evaluation for MRD in B-LL/L by flow cytometry relies on a knowledge of normal immunophenotypic patterns associated with B-cell maturation in states of rest and marrow regeneration so that one can identify patterns of antigen expression that differentiate abnormal, leukemic populations from regenerating hematogones or B-cell precursors. The nature of therapy can affect normal patterns, a phenomenon especially important to take into consideration given the increased use of targeted therapies in the treatment of B-LL/L. CONCLUSIONS Flow cytometry is widely available in many laboratories and is a cost-effective way to evaluate for B-LL/L MRD. However, panel validation and interpreter education are crucial for accurate assessment.
Collapse
Affiliation(s)
- Sindhu Cherian
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| | - Lorinda A Soma
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| |
Collapse
|
31
|
Herborg LL, Nederby L, Brøndum RF, Hansen M, Hokland P, Roug AS. Antigen Expression Varies Significantly between Molecular Subgroups of Acute Myeloid Leukemia Patients: Clinical Applicability Is Hampered by Establishment of Relevant Cutoffs. Acta Haematol 2020; 144:275-284. [PMID: 33271547 DOI: 10.1159/000510504] [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: 06/04/2020] [Accepted: 07/27/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION In this single-center study of 268 acute myeloid leukemia (AML) patients, we have tested if a subset of 4 routinely employed immunophenotypic stem cell-associated markers correlated with the presence of recurrently mutated genes and if the markers were predictive for mutational status. METHODS Immunophenotypic data from 268 diagnostic AML samples obtained in 2009-2018 were analyzed retrospectively for the antigens CD34, CD117, CD123, and CLEC12A. Correlation between immunophenotypes and mutations was analyzed by Fischer's exact test. Clinical applicability of the markers for predicting mutational status was evaluated by receiver operating characteristics analyses, where an area under the curve (AUC) of at least 0.85 was accepted as clinically relevant. RESULTS For a number of genes, the antigen expression differed significantly between mutated and wild-type gene expression. Despite low AUCs, CD123 and CLEC12A correlated with FLT3+NPM1- and FLT3+NPM1+. Three subsets met the AUC requirements (CD34+, CD34+CD117+, and CD34-CD117+) for predicting FLT3-NPM1+ or FLT3+NPM1+. CONCLUSION The value of immunophenotypes as surrogate markers for mutational status in AML seems limited when employing CD123 and CLEC12A in combination with CD34 and CD117. Defining relevant cutoffs for given markers is challenging and hampered by variation between laboratories and patient groups.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD34/genetics
- Antigens, CD34/metabolism
- Area Under Curve
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunophenotyping
- Interleukin-3 Receptor alpha Subunit/genetics
- Interleukin-3 Receptor alpha Subunit/metabolism
- Kaplan-Meier Estimate
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Male
- Middle Aged
- Mutation
- Nucleophosmin
- Proto-Oncogene Proteins c-kit/genetics
- Proto-Oncogene Proteins c-kit/metabolism
- ROC Curve
- Receptors, Mitogen/genetics
- Receptors, Mitogen/metabolism
- Retrospective Studies
- Young Adult
Collapse
Affiliation(s)
| | - Line Nederby
- Department of Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Rasmus Froberg Brøndum
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Maria Hansen
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Peter Hokland
- Department of Hematology, Aarhus University Hospital, Aarhus N, Denmark
| | - Anne Stidsholt Roug
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark,
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark,
| |
Collapse
|
32
|
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: 2.3] [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
|
33
|
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
|
34
|
Bewersdorf JP, Shallis RM, Boddu PC, Wood B, Radich J, Halene S, Zeidan AM. The minimal that kills: Why defining and targeting measurable residual disease is the “Sine Qua Non” for further progress in management of acute myeloid leukemia. Blood Rev 2020; 43:100650. [DOI: 10.1016/j.blre.2019.100650] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
|
35
|
Salas MQ, Chen S, Lam W, Pasic I, Gerbitz A, Michelis FV, Kim D(DH, Al-Shaibani Z, Lipton JH, Mattsson J, Kumar R, Viswabandya A, Law AD. Less Is More: Superior Graft-versus-Host Disease-Free/Relapse-Free Survival with Reduced-Intensity Conditioning and Dual T Cell Depletion in Acute Myelogenous Leukemia. Biol Blood Marrow Transplant 2020; 26:1511-1519. [DOI: 10.1016/j.bbmt.2020.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/16/2020] [Accepted: 04/18/2020] [Indexed: 01/22/2023]
|
36
|
Kim IS. Minimal residual disease in acute lymphoblastic leukemia: technical aspects and implications for clinical interpretation. Blood Res 2020; 55:S19-S26. [PMID: 32719172 PMCID: PMC7386891 DOI: 10.5045/br.2020.s004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
Minimal residual disease (MRD) monitoring has proven to be one of the fundamental independent prognostic factors for patients with acute lymphoblastic leukemia (ALL). Sequential monitoring of MRD using sensitive and specific methods, such as real-time quantitative polymerase chain reaction (qPCR) or flow cytometry (FCM), has improved the assessment of treatment response and is currently used for therapeutic stratification and early detection. Although both FCM and qPCR yield highly consistent results with sensitivities of 10‒4, each method has several limitations. For example, qPCR is time-consuming and laborious: designing primers that correspond to the immunoglobulin (IG) and T-cell receptor (TCR) gene rearrangements at diagnosis can take 3‒4 weeks. In addition, the evolution of additional clones beyond the first or index clone during therapy cannot be detected, which might lead to false-negative results. FCM requires experienced technicians and sometimes does not achieve a sensitivity of 10‒4. Accordingly, a next generation sequencing (NGS)-based method has been developed in an attempt to overcome these limitations. With the advent of high-throughput NGS technologies, a more in-depth analysis of IG and/or TCR gene rearrangements is now within reach, which impacts all applications of IG/TR analysis. However, standardization, quality control, and validation of this new technology are warranted prior to its incorporation into routine practice.
Collapse
Affiliation(s)
- In-Suk Kim
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
37
|
Zhang R, Zhu H, Yuan Y, Zhao J, Yang X, Tian Z. Risk Factors for Relapse of Childhood B Cell Acute Lymphoblastic Leukemia. Med Sci Monit 2020; 26:e923271. [PMID: 32619211 PMCID: PMC7353297 DOI: 10.12659/msm.923271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background B cell acute lymphoblastic leukemia (B-ALL) is the most common type of ALL. This study aimed to explore risk factors for relapse of childhood B-ALL. Material/Methods Total of 102 pediatric B-ALL patients were included in this study. B-ALL patients were divided into a relapse group and a non-relapse group. Chemotherapy-induced agranulocytosis time, fusion gene, and minimal residual disease (MRD) were assessed. White blood cell (WBC) count in peripheral blood and risk stratification were evaluated in newly-diagnosed patients. Kaplan-Meier plots were used to evaluate the correlation between risk factors and relapse rates. Multivariate analysis was performed with Cox proportional hazard model to estimate relative risk (RR), 95% confidence interval (95% CI), and hazard ratio (HR). Finally, 99 cases of B-ALL were included in this study. Results There were significant differences between the relapse group and the non-relapse group in age (p=0.004), chemotherapy-induced agranulocytopenia (p=0.001), WBC count in peripheral blood of newly diagnosed patients (p=0.016), risk stratification (p=0.000), and MRD at 12th week (p=0.007). Age over 10 years, high-risk stratification, long period of agranulocytopenia, higher WBC counts, and MRD more than 10−4 were correlated with higher B-ALL relapse rate (p<0.05). Multivariate analysis showed significantly higher relapse rates for age ≥10 years, high-risk stratification, and MRD at 12th week >10−4, with RR (95% CI) of 4.001 (1.005–15.930), 4.964 (1.050–23.456), and 4.646 (1.383–15.614), respectively. Conclusions Agranulocytopenia ≤7 days, peripheral blood WBC >100×109/L, and MRD at 33rd day >10−4 were associated with B-ALL relapse. Age ≥10 years, high-risk stratification, and MRD at 12th week >10−4 were independent risk factors for relapse.
Collapse
Affiliation(s)
- Rongrong Zhang
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Haiyan Zhu
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Yufang Yuan
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Jiou Zhao
- Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu, China (mainland)
| | - Xiaochun Yang
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Zhaofang Tian
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| |
Collapse
|
38
|
Jacqmin H, Chatelain B, Louveaux Q, Jacqmin P, Dogné JM, Graux C, Mullier F. Clustering and Kernel Density Estimation for Assessment of Measurable Residual Disease by Flow Cytometry. Diagnostics (Basel) 2020; 10:E317. [PMID: 32443428 PMCID: PMC7277972 DOI: 10.3390/diagnostics10050317] [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: 03/26/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/04/2022] Open
Abstract
Standardization, data mining techniques, and comparison to normality are changing the landscape of multiparameter flow cytometry in clinical hematology. On the basis of these principles, a strategy was developed for measurable residual disease (MRD) assessment. Herein, suspicious cell clusters are first identified at diagnosis using a clustering algorithm. Subsequently, automated multidimensional spaces, named "Clouds", are created around these clusters on the basis of density calculations. This step identifies the immunophenotypic pattern of the suspicious cell clusters. Thereafter, using reference samples, the "Abnormality Ratio" (AR) of each Cloud is calculated, and major malignant Clouds are retained, known as "Leukemic Clouds" (L-Clouds). In follow-up samples, MRD is identified when more cells fall into a patient's L-Cloud compared to reference samples (AR concept). This workflow was applied on simulated data and real-life leukemia flow cytometry data. On simulated data, strong patient-dependent positive correlation (R2 = 1) was observed between the AR and spiked-in leukemia cells. On real patient data, AR kinetics was in line with the clinical evolution for five out of six patients. In conclusion, we present a convenient flow cytometry data analysis approach for the follow-up of hematological malignancies. Further evaluation and validation on more patient samples and different flow cytometry panels is required before implementation in clinical practice.
Collapse
Affiliation(s)
- Hugues Jacqmin
- Hematology Laboratory, NAmur Research Institute for LIfe Sciences (NARILIS), Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université catholique de Louvain, 5530 Yvoir, Belgium; (B.C.); (F.M.)
| | - Bernard Chatelain
- Hematology Laboratory, NAmur Research Institute for LIfe Sciences (NARILIS), Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université catholique de Louvain, 5530 Yvoir, Belgium; (B.C.); (F.M.)
| | | | | | | | - Carlos Graux
- Department of Hematology, Namur Research Institute for Life Sciences (NARILIS), Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université catholique de Louvain, 5530 Yvoir, Belgium;
| | - François Mullier
- Hematology Laboratory, NAmur Research Institute for LIfe Sciences (NARILIS), Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université catholique de Louvain, 5530 Yvoir, Belgium; (B.C.); (F.M.)
| |
Collapse
|
39
|
Rathe M, Preiss B, Marquart HV, Schmiegelow K, Wehner PS. Minimal residual disease monitoring cannot fully replace bone marrow morphology in assessing disease status in pediatric acute lymphoblastic leukemia. APMIS 2020; 128:414-419. [PMID: 32108963 DOI: 10.1111/apm.13037] [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: 10/28/2019] [Accepted: 02/24/2019] [Indexed: 01/24/2023]
Abstract
Minimal residual disease (MRD) monitoring has a strong prognostic value in childhood lymphoblastic leukemia (ALL) and is currently utilized in all major pediatric ALL protocols. MRD monitoring is done by multiparameter flow cytometry, IG/TCR quantitative PCR or reverse transcriptase quantitative PCR of leukemic fusion transcripts providing a reliable measurement of treatment response. However, occasionally bone marrow (BM) aspirates may not yield representative material or be misinterpreted due to treatment-induced changes in MRD marker profile, undetected subclones at diagnosis, contamination with peripheral blood or cell adhesion and stroma cell interactions posing a risk for underestimating MRD levels and misclassifying resistant disease that may be detected by traditional BM morphology methods, immunohistochemistry, karyotyping and FISH. We present four cases with high MRD levels where MRD monitoring failed to provide the correct stratification information. Through these cases, we discuss the continued need to consider all available information including BM smears, touch imprints and trephine biopsy preparations not only at diagnosis but throughout remission monitoring in pediatric ALL.
Collapse
Affiliation(s)
- Mathias Rathe
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Birgitte Preiss
- Department of Pathology, Research Unit of Pathology, Odense University Hospital, Odense, Denmark
| | - Hanne Vibeke Marquart
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Peder Skov Wehner
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| |
Collapse
|
40
|
Kruse A, Abdel-Azim N, Kim HN, Ruan Y, Phan V, Ogana H, Wang W, Lee R, Gang EJ, Khazal S, Kim YM. Minimal Residual Disease Detection in Acute Lymphoblastic Leukemia. Int J Mol Sci 2020; 21:E1054. [PMID: 32033444 PMCID: PMC7037356 DOI: 10.3390/ijms21031054] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 02/04/2023] Open
Abstract
Minimal residual disease (MRD) refers to a chemotherapy/radiotherapy-surviving leukemia cell population that gives rise to relapse of the disease. The detection of MRD is critical for predicting the outcome and for selecting the intensity of further treatment strategies. The development of various new diagnostic platforms, including next-generation sequencing (NGS), has introduced significant advances in the sensitivity of MRD diagnostics. Here, we review current methods to diagnose MRD through phenotypic marker patterns or differential gene patterns through analysis by flow cytometry (FCM), polymerase chain reaction (PCR), real-time quantitative polymerase chain reaction (RQ-PCR), reverse transcription polymerase chain reaction (RT-PCR) or NGS. Future advances in clinical procedures will be molded by practical feasibility and patient needs regarding greater diagnostic sensitivity.
Collapse
Affiliation(s)
- Aaron Kruse
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - Nour Abdel-Azim
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - Hye Na Kim
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - Yongsheng Ruan
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - Valerie Phan
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - Heather Ogana
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - William Wang
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - Rachel Lee
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - Eun Ji Gang
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| | - Sajad Khazal
- Department of Pediatrics Patient Care, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yong-Mi Kim
- Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Boulevard, MS #57, Los Angeles, CA 90027, USA; (A.K.); (N.A.-A.); (H.N.K.); (Y.R.); (V.P.); (H.O.); (W.W.); (R.L.); (E.J.G.)
| |
Collapse
|
41
|
Camburn AE, Petrasich M, Ruskova A, Chan G. Re: Myeloblasts in normal bone marrows expressing leukaemia-associated immunophenotypes: author reply. Pathology 2020; 52:291. [PMID: 31928759 DOI: 10.1016/j.pathol.2019.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 11/19/2019] [Indexed: 11/30/2022]
Affiliation(s)
| | | | - Anna Ruskova
- LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - George Chan
- LabPlus, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
42
|
Feng J, Chen XJ, Liu XM, Zou Y, Guo Y, Yang WY, Chen YM, Zhang L, Wang SC, Ruan M, Liu F, Liu TF, Qi BQ, Zhu XF, Wang HJ. [A correlation study between the minimal residual disease detection using multiparameter flow cytometry and prognosis of childhood acute B lymphoblastic leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:678-680. [PMID: 31495136 PMCID: PMC7342875 DOI: 10.3760/cma.j.issn.0253-2727.2019.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Indexed: 11/17/2022]
Affiliation(s)
- J Feng
- Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Della Starza I, Chiaretti S, De Propris MS, Elia L, Cavalli M, De Novi LA, Soscia R, Messina M, Vitale A, Guarini A, Foà R. Minimal Residual Disease in Acute Lymphoblastic Leukemia: Technical and Clinical Advances. Front Oncol 2019; 9:726. [PMID: 31448230 PMCID: PMC6692455 DOI: 10.3389/fonc.2019.00726] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
Introduction: Acute lymphoblastic leukemia (ALL) is the first neoplasm where the assessment of early response to therapy by minimal residual disease (MRD) monitoring has proven to be a fundamental tool to guide therapeutic choices. The most standardized methods to study MRD in ALL are multi-parametric flow cytometry (MFC) and polymerase chain reaction (PCR) amplification-based methods. Emerging technologies hold the promise to improve MRD detection in ALL patients. Moreover, novel therapies, such as monoclonal antibodies, bispecific T-cell engagers, and chimeric antigen receptor T cells (CART) represent exciting advancements in the management of B-cell precursor (BCP)-ALL. Aims: Through a review of the literature and in house data, we analyze the current status of MRD assessment in ALL to better understand how some of its limitations could be overcome by emerging molecular technologies. Furthermore, we highlight the future role of MRD monitoring in the context of personalized protocols, taking into account the genetic complexity in ALL. Results and Conclusions: Molecular rearrangements (gene fusions and immunoglobulin and T-cell receptor-IG/TR gene rearrangements) are widely used as targets to detect residual leukemic cells in ALL patients. The advent of novel techniques, namely next generation flow cytometry (NGF), digital-droplet-PCR (ddPCR), and next generation sequencing (NGS) appear important tools to evaluate MRD in ALL, since they have the potential to overcome the limitations of standard approaches. It is likely that in the forthcoming future these techniques will be incorporated in clinical trials, at least at decisional time points. Finally, the advent of new powerful compounds is further increasing MRD negativity rates, with benefits in long-term survival and a potential reduction of therapy-related toxicities. However, the prognostic relevance in the setting of novel immunotherapies still needs to be evaluated.
Collapse
Affiliation(s)
- Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,GIMEMA Foundation, Rome, Italy
| | - Sabina Chiaretti
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria S De Propris
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Loredana Elia
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia A De Novi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Monica Messina
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonella Vitale
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Guarini
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
44
|
Abstract
Increasing evidence supports the prognostic significance of measurable residual disease (MRD) in acute myeloid leukemia (AML). Dynamic MRD assessment for patients with AML complements baseline patient risk assessment factors in determining patient prognosis. MRD status may also be helpful in informing therapeutic decisions. The European Leukemia Net MRD working party recently issued consensus recommendations for the use of MRD in AML. The Food and Drug Administration also issued advice for using MRD in trials of hematologic malignancies. This article discusses MRD testing, highlights the challenges in adopting MRD testing in clinical practice, and provides insights into the future of the field.
Collapse
|
45
|
Affiliation(s)
- Anna Porwit
- Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of MedicineLund University Lund Sweden
| | | |
Collapse
|
46
|
Affiliation(s)
- Anna Porwit
- Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of MedicineLund University Lund Sweden
| | | |
Collapse
|
47
|
Porwit A, Béné MC. Multiparameter flow cytometry applications in the diagnosis of mixed phenotype acute leukemia. CYTOMETRY PART B-CLINICAL CYTOMETRY 2019. [DOI: 10.1002/cyto.b.21783 or extractvalue(1224,concat(0x5c,0x7170707871,(select (elt(1224=1224,1))),0x7162627671))] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Anna Porwit
- Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of MedicineLund University Lund Sweden
| | | |
Collapse
|
48
|
Affiliation(s)
- Anna Porwit
- Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of MedicineLund University Lund Sweden
| | | |
Collapse
|
49
|
Affiliation(s)
- Anna Porwit
- Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of MedicineLund University Lund Sweden
| | | |
Collapse
|
50
|
Porwit A, Béné MC. Multiparameter flow cytometry applications in the diagnosis of mixed phenotype acute leukemia. CYTOMETRY PART B-CLINICAL CYTOMETRY 2019. [DOI: 10.1002/cyto.b.21783 rlike (select (case when (2397=1595) then 0x31302e313030322f6379746f2e622e3231373833 else 0x28 end))-- mlwg] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Anna Porwit
- Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of MedicineLund University Lund Sweden
| | | |
Collapse
|