1
|
Danylesko I, Shem-Tov N, Yerushalmi R, Jacoby E, Toren A, Shouval R, Itzhaki O, Avigdor A, Shimoni A, Nagler A. Point of care CD19 chimeric antigen receptor (CAR) T-cells for relapsed/refractory acute myeloid leukemia (AML) with aberrant CD19 antigen expression. Curr Res Transl Med 2024; 72:103471. [PMID: 39305562 DOI: 10.1016/j.retram.2024.103471] [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: 05/09/2024] [Revised: 08/10/2024] [Accepted: 09/14/2024] [Indexed: 12/07/2024]
Abstract
Relapsed/refractory (r/r) acute myeloid leukemia (AML) is associated with poor prognosis. CD19 is a B-cell marker, is aberrantly expressed in AML, mostly with t(8; 21)(q22; q22.1). Here we report the results of a phase 2 study giving point of care produced CD19 CAR T- cells for r/r AML with aberrant expression of CD19 (NCT04257175). Lymphodepletion included fludarabine and cyclophosphamide The response was evaluated by bone marrow (BM) aspiration on day 28. Six patients (5 adults and 1 child) were included. Median number of previous chemotherapy lines was 4 (range, 3-8) and four patients received CAR T-cells 8-18 months post allogeneic hematopoietic stem cell transplantation (allo-HSCT). Cytokine release syndrome (CRS) of any grade occurred in all patients, and 1 patient had grade 3 CRS. Immune effector cell-associated neurotoxicity syndrome (ICANS) occurred in 2 patients at low grades. Tocilizumab was administered to 2 patients and corticosteroids to 3 patients. Four patients achieved a complete remission (CR), while 2/6 progressed (PD). Three patients (2 with CR and 1 with PD) underwent allo-HSCT (it was the second transplant in 2) 2-5 months post CAR T-cells infusion. The median duration of response in patients achieving CR was 8.5 (range; 3-14) months. However, all patients eventually died within 5 (1-18) months. In conclusion, CD19 CAR T- cell treatment for AML is feasible and safe. However, the response is short and should be followed by allo-HSCT. Hopefully, future long term results will be improved by combining the CAR T- cell therapy with the emerging novel effective anti-leukemic compounds.
Collapse
Affiliation(s)
- Ivetta Danylesko
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Noga Shem-Tov
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Yerushalmi
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elad Jacoby
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Amos Toren
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Roni Shouval
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Department of Medicine, Weill Cornell Medical College, New York, New York, USA; Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Orit Itzhaki
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Abraham Avigdor
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avichai Shimoni
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arnon Nagler
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
2
|
Huang P, Zhang C, Zhang A, Mao J, Liu G, Hu C, Zhu H. CD56 briCD38 + as a novel neutrophil-specific marker in chronic myeloid leukemia. Heliyon 2024; 10:e39465. [PMID: 39559206 PMCID: PMC11570290 DOI: 10.1016/j.heliyon.2024.e39465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Background Chronic myeloid leukemia (CML) is classified as a subtype of myeloproliferative neoplasm, and bone marrow flow cytometry does not reveal any specific immunophenotype. Interestingly, aberrant expression of cluster of differentiation (CD) markers such as CD56 and CD38 has been observed on neutrophils in CML patients. Therefore, we investigated the abnormal expression of CD56 and CD38 in CML neutrophils to explore their diagnostic value in identifying CML through flow cytometry. Methods We have developed a multi-parameter flow cytometry assay to identify aberrant immunophenotypes in CML neutrophils among bone marrow nucleated cells. Results Compared to healthy donors and patients with a reactive neutrophilia or other hematological malignancies, the percentage of CD56briCD38+ neutrophil subsets in CML patients exhibits a distinctive increase (cut-off value, 2.0 %). The specificity and sensitivity associated with the learning cohort (168 samples) were 90.8 % and 84.9 %, respectively, while in the validation cohort (194 samples), they were 90.7 % and 84.7 %. The accumulation of CD56briCD38+ neutrophil subsets, which demonstrate are abnormal characteristics, is independent of the neutrophil count, BCR/ABL1 fusions and risk stratification but associated with blast cells immunophenotype. Moreover, this increase disappears in CML patients after treatment with tyrosine kinase inhibitors when the curative effect was satisfactory. Conclusions We conclude that an increase in the proportion of CD56briCD38+ neutrophil subsets exceeding 2.0 % of total neutrophils serves as a highly sensitive and specific flow cytometry marker, enabling rapid and accurate identification of CML.
Collapse
Affiliation(s)
- Panpan Huang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Cuiping Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Aimei Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Ju Mao
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Gan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Chaojie Hu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Huaiping Zhu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| |
Collapse
|
3
|
Verigou E, Chatzilygeroudi T, Lazaris V, de Lastic AL, Symeonidis A. Immunophenotyping myelodysplastic neoplasms: the role of flow cytometry in the molecular classification era. Front Oncol 2024; 14:1447001. [PMID: 39544295 PMCID: PMC11560873 DOI: 10.3389/fonc.2024.1447001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
The unique heterogenous landscape of myelodysplastic syndromes/neoplasms (MDS) has resulted in continuous redefinition of disease sub-entities, in view of the novel translational research data that have clarified several areas of the pathogenesis and the progression of the disease. The new international classifications (WHO 2022, ICC 2022) have incorporated genomic data defining phenotypical alterations, that guide clinical management of specific patient subgroups. On the other hand, for over a decade, multiparameter flow cytometry (MFC) has proven its value as a complementary diagnostic tool for these diseases and although it has never been established as a mandatory test for the baseline evaluation of MDS patients in international guidelines, it is almost universally adopted in everyday clinical practice for the assessment of suspected cytopenias through simplified scoring systems or elaborate analytical strategies for the detection of immunophenotypical dysplastic features in every hematopoietic cell lineage in the bone marrow (BM). In this review, we explore the clinically meaningful interplay of MFC data and genetic profiles of MDS patients, to reveal the currently existing and the potential future role of each methodology for routine clinical practice, and the benefit of the patients. We reviewed the existing knowledge and recent advances in the field and discuss how an integrated approach could lead to patient re-stratification and guide personalized management.
Collapse
Affiliation(s)
- Evgenia Verigou
- Hematology Division, Department of Internal Medicine, General University Hospital of Patras - School of Medicine, Patras, Greece
| | - Theodora Chatzilygeroudi
- Hematology Division, Department of Internal Medicine, General University Hospital of Patras - School of Medicine, Patras, Greece
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | | | - Anne-Lise de Lastic
- Laboratory of Immunohematology, School of Medicine, University of Patras, Patras, Greece
| | - Argiris Symeonidis
- Hematology Division, Department of Internal Medicine, General University Hospital of Patras - School of Medicine, Patras, Greece
| |
Collapse
|
4
|
Shaforostova I, Call S, Evers G, Reicherts C, Angenendt L, Stelljes M, Berdel WE, Pohlmann A, Mikesch J, Rosenbauer F, Lenz G, Schliemann C, Wethmar K. Prevalence and clinical impact of CD56 and T-cell marker expression in acute myeloid leukaemia: A single-centre retrospective analysis. EJHAEM 2024; 5:93-104. [PMID: 38406551 PMCID: PMC10887264 DOI: 10.1002/jha2.827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 02/27/2024]
Abstract
Flow cytometry-based immunophenotyping is a mainstay of diagnostics in acute myeloid leukaemia (AML). Aberrant CD56 and T-cell antigen expression is observed in a fraction subset of AML cases, but the clinical relevance remains incompletely understood. Here, we retrospectively investigated the association of CD56 and T-cell marker expression with disease-specific characteristics and outcome of 324 AML patients who received intensive induction therapy at our centre between 2011 and 2019. We found that CD2 expression was associated with abnormal non-complex karyotype, NPM1 wild-type status and TP53 mutation. CD2 also correlated with a lower complete remission (CR) rate (47.8% vs. 71.6%, p = 0.03). CyTdT and CD2 were associated with inferior 3-year event-free-survival (EFS) (5.3% vs. 33.5%, p = 0.003 and 17.4% vs. 33.1%, p = 0.02, respectively). CyTdT expression was also correlated with inferior relapse-free survival (27.3% vs. 48.8%, p = 0.04). In multivariable analyses CD2 positivity was an independent adverse factor for EFS (HR 1.72, p = 0.03). These results indicate a biological relevance of aberrant T-cell marker expression in AML and provide a rationale to further characterise the molecular origin in T-lineage-associated AML.
Collapse
Affiliation(s)
| | - Simon Call
- Department of Medicine AUniversity Hospital MünsterMünsterGermany
| | - Georg Evers
- Department of Medicine AUniversity Hospital MünsterMünsterGermany
| | | | - Linus Angenendt
- Department of Medicine AUniversity Hospital MünsterMünsterGermany
- Department of Biosystems Science and EngineeringETH ZurichZürichSwitzerland
| | | | | | | | | | - Frank Rosenbauer
- Institute of Molecular Tumor BiologyFaculty of MedicineUniversity of MünsterMünsterGermany
| | - Georg Lenz
- Department of Medicine AUniversity Hospital MünsterMünsterGermany
| | | | - Klaus Wethmar
- Department of Medicine AUniversity Hospital MünsterMünsterGermany
| |
Collapse
|
5
|
Garg S, Ni W, Griffin JD, Sattler M. Chimeric Antigen Receptor T Cell Therapy in Acute Myeloid Leukemia: Trials and Tribulations. Hematol Rep 2023; 15:608-626. [PMID: 37987319 PMCID: PMC10660693 DOI: 10.3390/hematolrep15040063] [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: 07/18/2023] [Revised: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy that is often associated with relapse and drug resistance after standard chemotherapy or targeted therapy, particularly in older patients. Hematopoietic stem cell transplants are looked upon as the ultimate salvage option with curative intent. Adoptive cell therapy using chimeric antigen receptors (CAR) has shown promise in B cell malignancies and is now being investigated in AML. Initial clinical trials have been disappointing in AML, and we review current strategies to improve efficacy for CAR approaches. The extensive number of clinical trials targeting different antigens likely reflects the genetic heterogeneity of AML. The limited number of patients reported in multiple early clinical studies makes it difficult to draw conclusions about CAR safety, but it does suggest that the efficacy of this approach in AML lags behind the success observed in B cell malignancies. There is a clear need not only to improve CAR design but also to identify targets in AML that show limited expression in normal myeloid lineage cells.
Collapse
Affiliation(s)
- Swati Garg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Ni
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - James D. Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Tettero JM, Dakappagari N, Heidinga ME, Oussoren-Brockhoff Y, Hanekamp D, Pahuja A, Burns K, Kaur P, Alfonso Z, van der Velden VHJ, Te Marvelde JG, Hobo W, Slomp J, Bachas C, Kelder A, Nguyen K, Cloos J. Analytical assay validation for acute myeloid leukemia measurable residual disease assessment by multiparametric flow cytometry. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:426-439. [PMID: 37766649 DOI: 10.1002/cyto.b.22144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Measurable residual disease (MRD) assessed by multiparametric flow cytometry (MFC) has gained importance in clinical decision-making for acute myeloid leukemia (AML) patients. However, complying with the recent In Vitro Diagnostic Regulations (IVDR) in Europe and Food and Drug Administration (FDA) guidance in the United States requires rigorous validation prior to their use in investigational clinical trials and diagnostics. Validating AML MRD-MFC assays poses challenges due to the unique underlying disease biology and paucity of patient specimens. In this study, we describe an experimental framework for validation that meets regulatory expectations. METHODS Our validation efforts focused on evaluating assay accuracy, analytical specificity, analytical and functional sensitivity (limit of blank (LoB), detection (LLoD) and quantitation (LLoQ)), precision, linearity, sample/reagent stability and establishing the assay background frequencies. RESULTS Correlation between different MFC methods was highly significant (r = 0.99 for %blasts and r = 0.93 for %LAIPs). The analysis of LAIP specificity accurately discriminated from negative control cells. The assay demonstrated a LoB of 0.03, LLoD of 0.04, and LLoQ of 0.1%. Precision experiments yielded highly reproducible results (Coefficient of Variation <20%). Stability experiments demonstrated reliable measurement of samples up to 96 h from collection. Furthermore, the reference range of LAIP frequencies in non-AML patients was below 0.1%, ranging from 0.0% to 0.04%. CONCLUSION In this manuscript, we present the validation of an AML MFC-MRD assay using BM/PB patient specimens, adhering to best practices. Our approach is expected to assist other laboratories in expediting their validation activities to fulfill recent health authority guidelines.
Collapse
Affiliation(s)
- Jesse M Tettero
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | | | - Maaike E Heidinga
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Yvonne Oussoren-Brockhoff
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Diana Hanekamp
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anil Pahuja
- Navigate BioPharma (a Novartis Subsidiary), Carlsbad, California, USA
| | - Kerri Burns
- Navigate BioPharma (a Novartis Subsidiary), Carlsbad, California, USA
| | - Pavinder Kaur
- Navigate BioPharma (a Novartis Subsidiary), Carlsbad, California, USA
| | - Zeni Alfonso
- Navigate BioPharma (a Novartis Subsidiary), Carlsbad, California, USA
| | | | - Jeroen G Te Marvelde
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine-Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jennichjen Slomp
- Department of Clinical Chemistry, Medisch Spectrum Twente/Medlon, Enschede, The Netherlands
| | - Costa Bachas
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Angele Kelder
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Kevin Nguyen
- Navigate BioPharma (a Novartis Subsidiary), Carlsbad, California, USA
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Albitar M, Zhang H, Charifa A, Ip A, Ma W, McCloskey J, Donato M, Siegel D, Waintraub S, Gutierrez M, Pecora A, Goy A. Combining cell-free RNA with cell-free DNA in liquid biopsy for hematologic and solid tumors. Heliyon 2023; 9:e16261. [PMID: 37251903 PMCID: PMC10208940 DOI: 10.1016/j.heliyon.2023.e16261] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/07/2023] [Accepted: 05/11/2023] [Indexed: 05/31/2023] Open
Abstract
Current use of liquid biopsy is based on cell-free DNA (cfDNA) and the evaluation of mutations or methylation pattern. However, expressed RNA can capture mutations, changes in expression levels due to methylation, and provide information on cell of origin, growth, and proliferation status. We developed an approach to isolate cell-free total nucleic acid (cfDNA) and used targeted next generation sequencing to sequence cell-free RNA (cfRNA) and cfDNA as new approach in liquid biopsy. We demonstrate that cfRNA is overall more sensitive than cfDNA in detecting mutations. We show that cfRNA is reliable in detecting fusion genes and cfDNA is reliable in detecting chromosomal gains and losses. cfRNA levels of various solid tumor biomarkers were significantly higher (P < 0.0001) in samples from solid tumors as compared with normal control. Similarly, cfRNA lymphoid markers and cfRNA myeloid markers were all higher in lymphoid and myeloid neoplasms, respectively as compared with control (P < 0.0001). Using machine learning we demonstrate cfRNA was highly predictive of diagnosis (AUC >0.98) of solid tumors, B-cell lymphoid neoplasms, T-cell lymphoid neoplasms, and myeloid neoplasms. In evaluating the host immune system, cfRNA CD4:CD8B and CD3D:CD19 ratios in normal controls were as expected (median: 5.92 and 6.87, respectively) and were significantly lower in solid tumors (P < 0.0002). This data suggests that liquid biopsy combining analysis of cfRNA with cfDNA is practical and may provide helpful information in predicting genomic abnormalities, diagnosis of neoplasms and evaluating both the tumor biology and the host response.
Collapse
Affiliation(s)
- Maher Albitar
- Genomic Testing Cooperative, LCA, Irvine, CA, 92618, USA
| | - Hong Zhang
- Genomic Testing Cooperative, LCA, Irvine, CA, 92618, USA
| | - Ahmad Charifa
- Genomic Testing Cooperative, LCA, Irvine, CA, 92618, USA
| | - Andrew Ip
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Wanlong Ma
- Genomic Testing Cooperative, LCA, Irvine, CA, 92618, USA
| | - James McCloskey
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Michele Donato
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - David Siegel
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Stanley Waintraub
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Martin Gutierrez
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Andrew Pecora
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Andre Goy
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| |
Collapse
|
8
|
Weeda V, Mestrum SGC, Leers MPG. Flow Cytometric Identification of Hematopoietic and Leukemic Blast Cells for Tailored Clinical Follow-Up of Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:ijms231810529. [PMID: 36142442 PMCID: PMC9506284 DOI: 10.3390/ijms231810529] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML) is a myeloid malignancy that is characterized by the accumulation of leukemic blast cells, which originate from hematopoietic stem cells that have undergone leukemic transformation and/or are more mature progenitors that have gained stemness features. Currently, no consensus exists for the flow cytometric identification of normal blast cells and their leukemic counterparts by their antigenic expression profile. Differentiating between the benign cells and the malignant cells is crucial for the further deployment of immunophenotype panels for the clinical follow-up of AML patients. This review provides an overview of immunophenotypic markers that allow the identification of leukemic blast cells in the bone marrow with multiparameter flow cytometry. This technique allows the identification of hematopoietic blast cells at the level of maturing cells by their antigen expression profile. While aberrant antigen expression of a single immunophenotypic marker cell cannot be utilized in order to differentiate leukemic blast cells from normal blast cells, combinations of multiple immunophenotypic markers can enable the distinction of normal and leukemic blast cells. The identification of these markers has provided new perspectives for tailored clinical follow-up, including therapy management, diagnostics, and prognostic purposes. The immunophenotypic marker panels, however, should be developed by carefully considering the variable antigen marker expression profile of individual patients.
Collapse
Affiliation(s)
- Vera Weeda
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
| | - Stefan G. C. Mestrum
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
- Department of Molecular Cell Biology, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, 6200MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-6-36176124
| | - Math P. G. Leers
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
| |
Collapse
|
9
|
Park SY, Suh KJ, Lee DW, Ryu HS, Kim M, Kim SH, Lee KH, Kim TY, Kim JH, Park IA, Im SA. Prognostic role of tumor subtype and germline BRCA mutation in advanced breast cancer patients treated with palbociclib plus endocrine therapy. Breast Cancer Res Treat 2022; 196:121-128. [PMID: 36070058 DOI: 10.1007/s10549-022-06566-8] [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: 08/13/2021] [Accepted: 03/12/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Palbociclib is a cyclin-dependent kinase 4 and 6 inhibitor which shows promising effect in hormone receptor-positive breast cancer. The purpose of this study is to evaluate the real-world efficacy and toxicity of palbociclib plus endocrine therapy. METHODS This is a retrospective study performed in two tertiary referral hospitals in Korea. Advanced breast cancer patients who were treated with 1st-line palbociclib plus endocrine therapy were enrolled. RESULTS A total of 216 patients were included between August 2016 and May 2019. Median age was 56 (29-89) years old and 75 patients (34.7%) were premenopausal. Median progression-free survival (PFS) was 33.0 months [95% confidence interval (CI) 24.7 to 41.3] and objective response rate was 59.3%. Luminal B patients had shorter PFS (33.0 months vs. Not reached, p = 0.019) and tendency of lower ORR (58.3 vs. 62.0%, p = 0.19) compared to luminal A patients. Multivariate analysis revealed luminal B (adjusted hazard ratio 1.90, p = 0.038) and germline BRCA mutation (adjusted hazard ratio 5.57, p = 0.002) as an independent poor prognostic factor for PFS. The most common grade 3 or 4 adverse event was neutropenia (86.7%). CONCLUSION The efficacy and toxicity of palbociclib in the real world were comparable to those of clinical trials. In addition, palbociclib with endocrine therapy was an effective treatment option for young patients. Luminal B and germline BRCA mutation were associated with inferior outcome.
Collapse
Affiliation(s)
- Song Yi Park
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Internal Medicine, Kangbuk Samsung Hospital, 29 Saemunan-ro, Jongno-gu, Seoul, 03181, Republic of Korea.,Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Koung Jin Suh
- Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, Gyeonggi-do, 13620, Republic of Korea
| | - Dae-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Han Suk Ryu
- Department of Pathology , Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Pathology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Cancer Research Institute, Seoul National University , 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Se Hyun Kim
- Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, Gyeonggi-do, 13620, Republic of Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Cancer Research Institute, Seoul National University , 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Cancer Research Institute, Seoul National University , 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jee Hyun Kim
- Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, Gyeonggi-do, 13620, Republic of Korea.,Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Cancer Research Institute, Seoul National University , 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
10
|
Piñero P, Morillas M, Gutierrez N, Barragán E, Such E, Breña J, García-Hernández MC, Gil C, Botella C, González-Navajas JM, Zapater P, Montesinos P, Sempere A, Tarín F. Identification of Leukemia-Associated Immunophenotypes by Databaseguided Flow Cytometry Provides a Highly Sensitive and Reproducible Strategy for the Study of Measurable Residual Disease in Acute Myeloblastic Leukemia. Cancers (Basel) 2022; 14:cancers14164010. [PMID: 36011002 PMCID: PMC9406948 DOI: 10.3390/cancers14164010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The complete immunophenotypic characterization of acute myeloid leukemia is essential for an accurate diagnosis and follow-up, which is determinant in the course of the disease. In many cases, the only option for the evaluation of minimal residual disease is flow cytometry, so the aim of this study is to develop an automatized multidimensional strategy to identify and characterize LAIPs as well as to detect new emerging aberrances in AML patients during the follow-up. The integrated DFN/LAIP strategy that we propose allows the identification of the most useful markers for minimal residual disease monitoring, improving the sensitivity and specificity of these studies. Furthermore, the use of databases and the automation of the analysis provide the basis for the generation of objective conclusions in minimal residual disease evaluations. Abstract Background: Multiparametric Flow Cytometry (MFC) is an essential tool to study the involved cell lineages, the aberrant differentiation/maturation patterns and the expression of aberrant antigens in acute myeloid leukemia (AML). The characterization of leukemia-associated immunophenotypes (LAIPs) at the moment of diagnosis is critical to establish reproducible strategies for the study of measurable residual disease using MFC (MFC-MRD). Methods: In this study, we identify and characterize LAIPs by comparing the leukemic populations of 145 AML patients, using the EuroFlow AML/ MDS MFC panel, with six databases of normal myeloid progenitors (MPCs). Principal component analysis was used to identify and characterize the LAIPs, which were then used to generate individual profiles for MFC-MRD monitoring. Furthermore, we investigated the relationship between the expression patterns of LAIPs and the different subtypes of AML. The MFC-MRD study was performed by identifying residual AML populations that matched with the LAIPs at diagnosis. To further validate this approach, the presence of MRD was also assessed by qPCR (qPCR-MRD). Finally, we studied the association between MFC-MRD and progression-free survival (PFS). Results: The strategy used in this study allowed us to describe more than 300 different LAIPs and facilitated the association of specific phenotypes with certain subtypes of AML. The MFC-MRD monitoring based on LAIPs with good/strong specificity was applicable to virtually all patients and showed a good correlation with qPCR-MRD and PFS. Conclusions: The described methodology provides an objective method to identify and characterize LAIPs. Furthermore, it provides a theoretical basis to develop highly sensitive MFC-MRD strategies.
Collapse
Affiliation(s)
- Paula Piñero
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
- Correspondence:
| | - Marina Morillas
- Hematology Department, Dr. Balmis General University Hospital, 03010 Alicante, Spain
| | - Natalia Gutierrez
- Hematology Department, Dr. Balmis General University Hospital, 03010 Alicante, Spain
| | - Eva Barragán
- Hematology Department, La Fe University Hospital, 46026 Valencia, Spain
| | - Esperanza Such
- Hematology Department, La Fe University Hospital, 46026 Valencia, Spain
| | - Joaquin Breña
- Hematology Department, La Candelaria General University Hospital, 38010 Santa Cruz de Tenerife, Spain
| | | | - Cristina Gil
- Hematology Department, Dr. Balmis General University Hospital, 03010 Alicante, Spain
| | - Carmen Botella
- Hematology Department, Dr. Balmis General University Hospital, 03010 Alicante, Spain
| | | | - Pedro Zapater
- Pharmacology Department, Dr. Balmis General University Hospital, Miguel Hernández University, 03202 Elche, Spain
| | - Pau Montesinos
- Hematology Department, La Fe University Hospital, 46026 Valencia, Spain
| | - Amparo Sempere
- Hematology Department, La Fe University Hospital, 46026 Valencia, Spain
| | - Fabian Tarín
- Hematology Department, Dr. Balmis General University Hospital, 03010 Alicante, Spain
| |
Collapse
|
11
|
Celant E, Marconato L, Stefanello D, Moretti P, Aresu L, Comazzi S, Martini V. Clinical and Clinical Pathological Presentation of 310 Dogs Affected by Lymphoma with Aberrant Antigen Expression Identified via Flow Cytometry. Vet Sci 2022; 9:vetsci9040184. [PMID: 35448684 PMCID: PMC9032799 DOI: 10.3390/vetsci9040184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Phenotypic aberrancies have been reported occasionally in canine lymphomas. Here, we retrospectively collected 310 canine lymphomas with an aberrant phenotype detected via flow cytometry and describe their clinical and clinical pathological features at diagnosis. There were 152 T-cell lymphomas not otherwise specified (T-NOS), 101 T-zone lymphomas (TZL), 54 B-cell lymphomas, and 3 cases with two suspected concurrent neoplastic populations. The most represented aberrancies were: CD5-, CD4-CD8-, and CD3- in T-NOS lymphomas, CD21+, CD4-CD8-, and CD3- in TZLs, and CD34+, CD44-, and CD5+ in B-cell lymphomas. Among T-cell lymphomas, the aberrant expression of CD21 was significantly more frequent in TZL and the loss of CD5 and CD44 in T-NOS. More than 75% of dogs were purebred; males outnumbered females; the mean age at diagnosis was 8–10 years, depending on lymphoma subtype. A few dogs were symptomatic at the time of diagnosis, and 30% had peripheral blood abnormalities, in line with what is already reported for the general population of dogs with lymphoma. Further studies are needed to assess the pathogenetic mechanisms underlying each specific antigen aberrancy, as well as the diagnostic and prognostic role.
Collapse
Affiliation(s)
- Elena Celant
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (E.C.); (D.S.); (P.M.); (S.C.)
| | - Laura Marconato
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, 40064 Bologna, Italy;
| | - Damiano Stefanello
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (E.C.); (D.S.); (P.M.); (S.C.)
| | - Pierangelo Moretti
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (E.C.); (D.S.); (P.M.); (S.C.)
| | - Luca Aresu
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, Grugliasco, 10095 Turin, Italy;
| | - Stefano Comazzi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (E.C.); (D.S.); (P.M.); (S.C.)
| | - Valeria Martini
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (E.C.); (D.S.); (P.M.); (S.C.)
- Correspondence: ; Tel.: +39-0250334585
| |
Collapse
|
12
|
Healy FM, Dahal LN, Jones JRE, Floisand Y, Woolley JF. Recent Progress in Interferon Therapy for Myeloid Malignancies. Front Oncol 2021; 11:769628. [PMID: 34778087 PMCID: PMC8586418 DOI: 10.3389/fonc.2021.769628] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/13/2021] [Indexed: 12/29/2022] Open
Abstract
Myeloid malignancies are a heterogeneous group of clonal haematopoietic disorders, caused by abnormalities in haematopoietic stem cells (HSCs) and myeloid progenitor cells that originate in the bone marrow niche. Each of these disorders are unique and present their own challenges with regards to treatment. Acute myeloid leukaemia (AML) is considered the most aggressive myeloid malignancy, only potentially curable with intensive cytotoxic chemotherapy with or without allogeneic haematopoietic stem cell transplantation. In comparison, patients diagnosed with chronic myeloid leukaemia (CML) and treated with tyrosine kinase inhibitors (TKIs) have a high rate of long-term survival. However, drug resistance and relapse are major issues in both these diseases. A growing body of evidence suggests that Interferons (IFNs) may be a useful therapy for myeloid malignancies, particularly in circumstances where patients are resistant to existing front-line therapies and have risk of relapse following haematopoietic stem cell transplant. IFNs are a major class of cytokines which are known to play an integral role in the non-specific immune response. IFN therapy has potential as a combination therapy in AML patients to reduce the impact of minimal residual disease on relapse. Alongside this, IFNs can potentially sensitize leukaemic cells to TKIs in resistant CML patients. There is evidence also that IFNs have a therapeutic role in myeloproliferative neoplasms (MPNs) such as polycythaemia vera (PV) and primary myelofibrosis (PMF), where they can restore polyclonality in patients. Novel formulations have improved the clinical effectiveness of IFNs. Low dose pegylated IFN formulations improve pharmacokinetics and improve patient tolerance to therapies, thereby minimizing the risk of haematological toxicities. Herein, we will discuss recent developments and the current understanding of the molecular and clinical implications of Type I IFNs for the treatment of myeloid malignancies.
Collapse
Affiliation(s)
- Fiona M Healy
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Lekh N Dahal
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Jack R E Jones
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Yngvar Floisand
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom.,The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - John F Woolley
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
13
|
Vonk CM, Al Hinai ASA, Hanekamp D, Valk PJM. Molecular Minimal Residual Disease Detection in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:5431. [PMID: 34771594 PMCID: PMC8582498 DOI: 10.3390/cancers13215431] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Initial induction chemotherapy to eradicate the bulk of acute myeloid leukemia (AML) cells results in complete remission (CR) in the majority of patients. However, leukemic cells persisting in the bone marrow below the morphologic threshold remain unaffected and have the potential to proliferate and re-emerge as AML relapse. Detection of minimal/measurable residual disease (MRD) is a promising prognostic marker for AML relapse as it can assess an individual patients' risk profile and evaluate their response to treatment. With the emergence of molecular techniques, such as next generation sequencing (NGS), a more sensitive assessment of molecular MRD markers is available. In recent years, the detection of MRD by molecular assays and its association with AML relapse and survival has been explored and verified in multiple studies. Although most studies show that the presence of MRD leads to a worse clinical outcome, molecular-based methods face several challenges including limited sensitivity/specificity, and a difficult distinction between mutations that are representative of AML rather than clonal hematopoiesis. This review describes the studies that have been performed using molecular-based assays for MRD detection in the context of other MRD detection approaches in AML, and discusses limitations, challenges and opportunities.
Collapse
Affiliation(s)
- Christian M Vonk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
| | - Adil S A Al Hinai
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
- National Genetic Center, Ministry of Health, Muscat 111, Oman
| | - Diana Hanekamp
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
- Department of Hematology, Cancer Center VU University Medical Center, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| | - Peter J M Valk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
| |
Collapse
|
14
|
Chashchina A, Märklin M, Hinterleitner C, Salih HR, Heitmann JS, Klimovich B. DNAM-1/CD226 is functionally expressed on acute myeloid leukemia (AML) cells and is associated with favorable prognosis. Sci Rep 2021; 11:18012. [PMID: 34504191 PMCID: PMC8429762 DOI: 10.1038/s41598-021-97400-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/20/2021] [Indexed: 11/09/2022] Open
Abstract
DNAM-1 is reportedly expressed on cytotoxic T and NK cells and, upon interaction with its ligands CD112 and CD155, plays an important role in tumor immunosurveillance. It has also been reported to be functionally expressed by myeloid cells, but expression and function on malignant cells of the myeloid lineage have not been studied so far. Here we analyzed expression of DNAM-1 in leukemic cells of acute myeloid leukemia (AML) patients. We found substantial levels of DNAM-1 to be expressed on leukemic blasts in 48 of 62 (> 75%) patients. Interaction of DNAM-1 with its ligands CD112 and CD155 induced release of the immunomodulatory cytokines IL-6, IL-8 IL-10 and TNF-α by AML cells and DNAM-1 expression correlated with a more differentiated phenotype. Multivariate analysis did not show any association of DNAM-1 positivity with established risk factors, but expression was significantly associated with clinical disease course: patients with high DNAM-1 surface levels had significantly longer progression-free and overall survival compared to DNAM-1low patients, independently whether patients had undergone allogenic stem cell transplantation or not. Together, our findings unravel a functional role of DNAM-1 in AML pathophysiology and identify DNAM-1 as a potential novel prognostic maker in AML.
Collapse
Affiliation(s)
- Anna Chashchina
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy (iFIT)", 72076, Tübingen, Germany
| | - Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy (iFIT)", 72076, Tübingen, Germany
| | - Clemens Hinterleitner
- DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy (iFIT)", 72076, Tübingen, Germany.,Department of Medical Oncology and Pulmonology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy (iFIT)", 72076, Tübingen, Germany
| | - Jonas S Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany. .,DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy (iFIT)", 72076, Tübingen, Germany.
| | - Boris Klimovich
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy (iFIT)", 72076, Tübingen, Germany
| |
Collapse
|
15
|
Hao Q, Liu X, Zhang Y, Zhang D, Li B, Wang J. MRD abnormal expression predict poor outcomes for refractory or relapsed acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. J Clin Lab Anal 2021; 35:e23974. [PMID: 34432318 PMCID: PMC8529132 DOI: 10.1002/jcla.23974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 12/16/2022] Open
Abstract
We retrospectively analyzed data from 197 patients with refractory or relapsed acute myeloid leukemia (r/rAML) who underwent allo‐HCT between January 2013 and February 2020 in our center (patients with promyelocytic leukemia were excluded). Of all patients, 86 achieved a complete morphological remission (CR) before transplant, while 111 failed to do so (NR). In the CR group, 32 patients displayed minimal residual disease (MRD‐positive). According to their immunophenotype pre‐HCT, we divided the MRD‐positive group and NR group into three subgroups: MRD 0+ group (without any antigen abnormal expression of CD7+, CD56+, CD38−, or HLA‐DR−) 28 patients, MRD 1+ group (with one abnormal antigen expression of CD7+, CD56+, CD38−, or HLA‐DR−) 63 patients, MRD 2+ group (with two or more abnormal antigens expression of CD7+, CD56+, CD38‐, or HLA‐DR‐) 52 patients. 3‐year estimates of disease‐free survival (DFS) for MRD 0+, MRD 1+ and MRD 2+ patients were 59.5 ± 9.5%, 29.9 ± 6.1%, and 9.4 ± 5.1%, and 3‐year estimates of overall survival (OS) were 59.5 ± 9.5%, 34.5 ± 6.3%, and 14.5 ± 10.8%, respectively. Multivariate analysis adjusted for genetic risk, blast cell level, secondary disease, age, sex, and donor relationship pre‐HCT, the hazard ratios of abnormal expression of CD7+, CD56+, HLA‐DR−, and CD38‐ were 6.69 (range 2.08–21.52; p = 0.001) for DFS, 2.24 (range 1.21–4.14; p = 0.010) for OS, and 7.18 (range 2.23–23.10; p = 0.001) for relapse compared with CD7−, CD56−, HLA‐DR+, and CD38+ patients. Our finding suggested that abnormal expression of CD7+, CD56+, HLA‐DR−, and CD38− is associated with poor outcomes, and the more number of abnormal antigens expression predict worse outcomes.
Collapse
Affiliation(s)
- Qi Hao
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Xinyue Liu
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Yongping Zhang
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Dongmei Zhang
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Boran Li
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Jingbo Wang
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
16
|
Hussein G, Jawad A. Impact of aberrant antigens expression on remission rate after first induction course of chemotherapy in de novo adult acute myeloid leukemia. IRAQI JOURNAL OF HEMATOLOGY 2021. [DOI: 10.4103/ijh.ijh_17_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
17
|
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
|
18
|
Daga S, Rosenberger A, Kashofer K, Heitzer E, Quehenberger F, Halbwedl I, Graf R, Krisper N, Prietl B, Höfler G, Reinisch A, Zebisch A, Sill H, Wölfler A. Sensitive and broadly applicable residual disease detection in acute myeloid leukemia using flow cytometry-based leukemic cell enrichment followed by mutational profiling. Am J Hematol 2020; 95:1148-1157. [PMID: 32602117 PMCID: PMC7540028 DOI: 10.1002/ajh.25918] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 12/19/2022]
Abstract
Persistent measurable residual disease (MRD) is an increasingly important prognostic marker in acute myeloid leukemia (AML). Currently, MRD is determined by multi-parameter flow cytometry (MFC) or PCR-based methods detecting leukemia-specific fusion transcripts and mutations. However, while MFC is highly operator-dependent and difficult to standardize, PCR-based methods are only available for a minority of AML patients. Here we describe a novel, highly sensitive and broadly applicable method for MRD detection by combining MFC-based leukemic cell enrichment using an optimized combinatorial antibody panel targeting CLL-1, TIM-3, CD123 and CD117, followed by mutational analysis of recurrently mutated genes in AML. In dilution experiments this method showed a sensitivity of 10-4 to 10-5 for residual disease detection. In prospectively collected remission samples this marker combination allowed for a median 67-fold cell enrichment with sufficient DNA quality for mutational analysis using next generation sequencing (NGS) or digital PCR in 39 out of 41 patients. Twenty-one samples (53.8%) tested MRD positive, whereas 18 (46.2%) were negative. With a median follow-up of 559 days, 71.4% of MRD positive (15/21) and 27.8% (5/18) of MRD negative patients relapsed (P = .007). The cumulative incidence of relapse (CIR) was higher for MRD positive patients (5-year CIR: 90.5% vs 28%, P < .001). In multivariate analysis, MRD positivity was a prominent factor for CIR. Thus, MFC-based leukemic cell enrichment using antibodies against CLL-1, TIM-3, CD123 and CD117 followed by mutational analysis allows high sensitive MRD detection and is informative on relapse risk in the majority of AML patients.
Collapse
Affiliation(s)
- Shruti Daga
- Division of HematologyMedical University of GrazGrazAustria
- CBmed Center of Biomarker Research in MedicineGrazAustria
| | | | - Karl Kashofer
- Division of PathologyMedical University of GrazGrazAustria
| | - Ellen Heitzer
- Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Franz Quehenberger
- Institute of Medical InformaticsStatistics and Documentation, Medical University of GrazGrazAustria
| | - Iris Halbwedl
- Division of PathologyMedical University of GrazGrazAustria
| | - Ricarda Graf
- Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Nina Krisper
- CBmed Center of Biomarker Research in MedicineGrazAustria
| | - Barbara Prietl
- CBmed Center of Biomarker Research in MedicineGrazAustria
| | - Gerald Höfler
- Division of PathologyMedical University of GrazGrazAustria
| | | | - Armin Zebisch
- Division of HematologyMedical University of GrazGrazAustria
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of PharmacologyMedical University of GrazGrazAustria
| | - Heinz Sill
- Division of HematologyMedical University of GrazGrazAustria
| | - Albert Wölfler
- Division of HematologyMedical University of GrazGrazAustria
- CBmed Center of Biomarker Research in MedicineGrazAustria
| |
Collapse
|
19
|
Panda D, Chatterjee G, Sardana R, Khanka T, Ghogale S, Deshpande N, Badrinath Y, Shetty D, Narula G, Banavali S, Patkar NV, Gujral S, Subramanian PG, Tembhare PR. Utility of CD36 as a novel addition to the immunophenotypic signature of RAM-phenotype acute myeloid leukemia and study of its clinicopathological characteristics. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:206-217. [PMID: 32865882 DOI: 10.1002/cyto.b.21943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/23/2020] [Accepted: 07/21/2020] [Indexed: 02/01/2023]
Abstract
INTRODUCTION In 2016, Children Oncology Group (COG) described a new high-risk subtype of acute myeloid leukemia (AML) with a distinct immunophenotypic-signature, RAM-phenotype (RAM-AML). Data on clinical and laboratory features of RAM-AML are still limited to COG report only. Herein, we report the clinicopathological characteristics and detailed immunophenotypic features of RAM-AML patients. In COG report, 38% of RAM-AML belonged to acute megakaryoblastic leukemia (AMKL)-subtype. Hence, we further compared the immunophenotypic features RAM-AML with non-RAM-AMKL diagnosed during the same study period. METHODS We included RAM-AML and non-RAM AMKL patients diagnosed between January 2017 and December 2019. We studied their morphological, cytochemical, immunophenotyping, cytogenetic, and molecular characteristics. Mean fluorescent intensity (MFI) and expression-pattern of immunophenotypic markers of RAM-AML were compared with non-RAM AMKLs patients. RESULTS We identified 11 RAM-AML (1%) and 21 non-RAM AMKL (1.9%) patients in 1102 pediatric-AML patients. Seven of 11 (63.64%) patients belonged to FAB-M7-subtype. CD56, CD117, and CD33 demonstrated overexpression, whereas CD45 and CD38 showed under-expression in RAM-AML patients. CD36 was consistently negative in RAM-AML, whereas moderate-bright positive in non-RAM AMKLs patients (p < 0.0001). On principle component analysis, addition of CD36 enhanced the visual-separation between RAM-AML and non-RAM AMKL clusters. Cytogenetic and molecular studies did not show any recurrent abnormality; however, RNA-sequencing study revealed CBFA2T3-GLIS2-fusion in three of seven (42.8%) RAM-AML patients. CONCLUSION We report the clinicopathological characteristics and the detailed immunophenotypic profile in the world's second series of RAM-AML patients. We further report a novel finding of CD36-negative expression as an additional parameter to the multidimensional immunophenotypic signature of this entity.
Collapse
Affiliation(s)
- Devasis Panda
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Gaurav Chatterjee
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Rohan Sardana
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Twinkle Khanka
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Sitaram Ghogale
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Nilesh Deshpande
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Yajamanam Badrinath
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Dhanalaxmi Shetty
- Department of Cancer Cytogenetics, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Gaurav Narula
- Department of Pediatric Oncology, Tata Memorial Center, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Shripad Banavali
- Department of Pediatric Oncology, Tata Memorial Center, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Nikhil V Patkar
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Sumeet Gujral
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Papagudi G Subramanian
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| | - Prashant R Tembhare
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, Maharashtra, India
| |
Collapse
|
20
|
Detection and management of acute myeloid leukemia measurable residual disease: is it standard of care? Curr Opin Hematol 2020; 27:81-87. [PMID: 31895104 DOI: 10.1097/moh.0000000000000566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW In the present manuscript, we will review the current approaches to investigate measurable residual disease (MRD) and its clinical applications in AML management. RECENT FINDINGS Over the last decades, several methods have been developed to trace MRD, with flow cytometry and polymerase chain reaction (PCR) being the most reliable. However, new technologies, such as digital PCR and Next-Generation Sequencing are emerging as particularly useful in AML. The 2017 European LeukemiaNet (ELN) recommendations have incorporated MRD assessment to define the response criteria to therapy, and more recently, the ELN MRD Working Party has published guidelines for the use of MRD in clinical practice. SUMMARY Morphologic complete remission (mCR) after induction therapy, has been consistently shown not only to have a critical prognostic role but also to fail in predicting relapse on an individual basis. Major attempts to improve our prediction capability have been made by measuring the residual levels of leukemic cells that persist in the bone marrow after chemotherapy. This number of cells, also called MRD, harbors in the bone marrow below the threshold of morphology and is responsible for leukemia recurrence. Therefore, the detection of MRD promises to help predict the risk of relapse, allowing a more proper patients' risk-stratification and the use of risk-tailored therapeutic strategy.
Collapse
|
21
|
Association of immunophenotype with expression of topoisomerase II α and β in adult acute myeloid leukemia. Sci Rep 2020; 10:5486. [PMID: 32218491 PMCID: PMC7099013 DOI: 10.1038/s41598-020-62345-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 03/12/2020] [Indexed: 01/13/2023] Open
Abstract
Anthracyclines used in the treatment of acute myelogenous leukemia (AML) inhibit the activity of the mammalian topoisomerase II (topo II) isoforms, topo II α and topo IIβ. In 230 patients with non-M3 AML who received frontline ara-C/daunorubicin we determined expression of topo IIα and topo IIβ by RT-PCR and its relationship to immunophenotype (IP) and outcomes. Treatment outcomes were analyzed by logistic or Cox regression. In 211 patients, available for analysis, topo IIα expression was significantly lower than topo IIβ (P < 0.0001). In contrast to topo IIα, topo IIβ was significantly associated with blast percentage in marrow or blood (P = 0.0001), CD7 (P = 0.01), CD14 (P < 0.0001) and CD54 (P < 0.0001). Event free survival was worse for CD56-negative compared to CD56-high (HR = 1.9, 95% CI [1.0-3.5], p = 0.04), and overall survival was worse for CD-15 low as compared to CD15-high (HR = 2.2, 95% CI [1.1-4.2], p = 0.02). Ingenuity pathway analysis indicated topo IIβ and immunophenotype markers in a network associated with cell-to-cell signaling, hematological system development/function and inflammatory response. Topo IIβ expression reflects disease biology of highly proliferative disease and distinct IP but does not appear to be an independent variable influencing outcome in adult AML patients treated with anthracycline-based therapy.
Collapse
|
22
|
A simple cytofluorimetric score may optimize testing for biallelic CEBPA mutations in patients with acute myeloid leukemia. Leuk Res 2019; 86:106223. [DOI: 10.1016/j.leukres.2019.106223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 11/24/2022]
|
23
|
Derman BA, Larson RA. Post-remission therapy in acute myeloid leukemia: Are we ready for an individualized approach? Best Pract Res Clin Haematol 2019; 32:101102. [PMID: 31779969 DOI: 10.1016/j.beha.2019.101102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Recent advances in remission induction treatment strategies for acute myeloid leukemia (AML) have improved the rates of complete remission (CR) and overall survival (OS), owing to a concerted effort to tailor therapies toward specific AML subtypes. However, without effective post-remission therapy, most patients will relapse. The extent to which post-remission therapies is individualized in the current paradigm is quite varied. Core binding factor (CBF) AML is typically treated with post-remission high-dose cytarabine (HiDAC) without allogeneic hematopoietic stem cell transplantation (HSCT), whereas those with intermediate or adverse-risk cytogenetics are treated with post-remission cytarabine followed by allogeneic HSCT in CR1 when feasible. A lack of clarity regarding the proper dosing of post-remission cytarabine has made consensus building on dosing and schedule a challenge. CBF AML benefits most from high-dose cytarabine (HiDAC), and dasatinib appears promising as an adjunct for those for KIT-mutated CBF AML. Other than series using CPX-351 or lomustine in older adults, multiagent chemotherapy approaches have resulted in excess toxicity without a survival benefit. Neither hypomethylating agents nor gemtuzumab ozogamicin have shown a material OS benefit. Targeted agents such as FLT3 inhibitors and IDH1/IDH2 inhibitors show potential for the patients who harbor these druggable targets, but few data are available. Many studies evaluating post-remission strategies to target AML in the MRD-positive state are already underway, and these remain a promising area of investigation.
Collapse
Affiliation(s)
- Benjamin A Derman
- Section of Hematology Oncology, Department of Medicine and Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA.
| | - Richard A Larson
- Section of Hematology Oncology, Department of Medicine and Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA
| |
Collapse
|
24
|
Transition from morphologic diagnosis to immunophenotypic diagnosis of acute leukemia—experience of establishing a new flow cytometry laboratory. J Hematop 2019. [DOI: 10.1007/s12308-019-00368-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
25
|
Ossenkoppele G, Schuurhuis GJ, van de Loosdrecht A, Cloos J. Can we incorporate MRD assessment into clinical practice in AML? Best Pract Res Clin Haematol 2019; 32:186-191. [DOI: 10.1016/j.beha.2019.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/08/2019] [Indexed: 12/29/2022]
|
26
|
Cummins KD, Gill S. Will CAR T cell therapy have a role in AML? Promises and pitfalls. Semin Hematol 2019; 56:155-163. [DOI: 10.1053/j.seminhematol.2018.08.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/27/2018] [Accepted: 08/27/2018] [Indexed: 12/27/2022]
|
27
|
Ehinger M, Pettersson L. Measurable residual disease testing for personalized treatment of acute myeloid leukemia. APMIS 2019; 127:337-351. [PMID: 30919505 DOI: 10.1111/apm.12926] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022]
Abstract
This review summarizes - with the practicing hematologist in mind - the methods used to determine measurable residual disease (MRD) in everyday practice with some future perspectives, and the current knowledge about the prognostic impact of MRD on outcome in acute myeloid leukemia (AML), excluding acute promyelocytic leukemia. Possible implications for choice of MRD method, timing of MRD monitoring, and guidance of therapy are discussed in general and in some detail for certain types of leukemia with specific molecular markers to monitor, including core binding factor (CBF)-leukemias and NPM1-mutated leukemias.
Collapse
Affiliation(s)
- Mats Ehinger
- Department of Clinical Sciences, Pathology, Skane University Hospital, Lund University, Lund, Sweden
| | - Louise Pettersson
- Department of Pathology, Halland Hospital Halmstad, Region Halland, Halmstad, Sweden.,Faculty of Medicine, Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Basharat M, Khan SA, Din NU, Ahmed D. Immunophenotypic characterisation of morphologically diagnosed cases of Acute Myeloid Leukaemia (AML). Pak J Med Sci 2019; 35:470-476. [PMID: 31086535 PMCID: PMC6500804 DOI: 10.12669/pjms.35.2.614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Objective: To determine immunophenotypic pattern in newly diagnosed cases of acute myeloid leukaemia by flow cytometry and its correlation with morphological findings. Methods: This study was conducted at Haematology (Pathology) department, Army Medical College, in collaboration with Immunology Department Armed Forces Institute of Pathology, Rawalpindi from 16 November 2016 to 16 November 2017. One hundred and six patients of both genders and all age groups diagnosed as acute myeloid leukaemia were included in the study. Demographic data was noted. Complete blood counts, bone marrow examination and cytochemical stains were carried out and evaluated microscopically for blast percentage and morphology. Immunophenotyping was performed by flow cytometry using standard panel on peripheral blood or bone marrow samples. The surface and cytoplasmic antigens of interest were analysed and correlated with morphological findings. Results: The most commonly expressed antigens were CD13, CD33, CD45 and HLA-DR. Almost all blasts expressed CD45 with no remarkable difference among the subtypes of AML. The mean positivity for CD13 among all AML subtypes was 57% and for CD33 was 67%. Aberrant expression of CD7 and CD19 were expressed in 26.4% and 1.1% of all cases respectively. There was concordance rate of 90% between morphology and FCM in our study. Conclusion: Flow cytometric analysis of acute leukaemia done by a combination of patterns and intensity of antigen expression improves diagnostic yield in AML. CD13, CD33 and CD45 are the most frequently expressed antigens in AML. Our findings suggest a 90% concordance between morphology and flow cytometry. It is pertinent to conclude that flow cytometry results interpreted with morphology are complementary.
Collapse
Affiliation(s)
- Maria Basharat
- Maria Basharat, MBBS. Pathology Department, Army Medical College, National University of Medical Sciences, Islamabad, Pakistan
| | - Saleem Ahmed Khan
- Saleem Ahmed Khan, MBBS, MCPS, FCPS, FRCP, PhD. Pathology Department, Army Medical College, National University of Medical Sciences, Islamabad, Pakistan
| | - Nasir Ud Din
- Nasir ud din MBBS, FCPS. Pathology Department, Army Medical College, National University of Medical Sciences, Islamabad, Pakistan
| | - Dawood Ahmed
- Dawood Ahmed MBBS, FCPS. Pathology Department, Army Medical College, National University of Medical Sciences, Islamabad, Pakistan
| |
Collapse
|
29
|
NCAM1 (CD56) promotes leukemogenesis and confers drug resistance in AML. Blood 2019; 133:2305-2319. [PMID: 30814062 DOI: 10.1182/blood-2018-12-889725] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023] Open
Abstract
Neural cell adhesion molecule 1 (NCAM1; CD56) is expressed in up to 20% of acute myeloid leukemia (AML) patients. NCAM1 is widely used as a marker of minimal residual disease; however, the biological function of NCAM1 in AML remains elusive. In this study, we investigated the impact of NCAM1 expression on leukemogenesis, drug resistance, and its role as a biomarker to guide therapy. Beside t(8;21) leukemia, NCAM1 expression was found in most molecular AML subgroups at highly heterogeneous expression levels. Using complementary genetic strategies, we demonstrated an essential role of NCAM1 in the regulation of cell survival and stress resistance. Perturbation of NCAM1 induced cell death or differentiation and sensitized leukemic blasts toward genotoxic agents in vitro and in vivo. Furthermore, Ncam1 was highly expressed in leukemic progenitor cells in a murine leukemia model, and genetic depletion of Ncam1 prolonged disease latency and significantly reduced leukemia-initiating cells upon serial transplantation. To further analyze the mechanism of the NCAM1-associated phenotype, we performed phosphoproteomics and transcriptomics in different AML cell lines. NCAM1 expression strongly associated with constitutive activation of the MAPK-signaling pathway, regulation of apoptosis, or glycolysis. Pharmacological inhibition of MEK1/2 specifically inhibited proliferation and sensitized NCAM1+ AML cells to chemotherapy. In summary, our data demonstrate that aberrant expression of NCAM1 is involved in the maintenance of leukemic stem cells and confers stress resistance, likely due to activation of the MAPK pathway. Targeting MEK1/2 sensitizes AML blasts to genotoxic agents, indicating a role for NCAM1 as a biomarker to guide AML treatment.
Collapse
|
30
|
Zhang T, de Waard AA, Wuhrer M, Spaapen RM. The Role of Glycosphingolipids in Immune Cell Functions. Front Immunol 2019; 10:90. [PMID: 30761148 PMCID: PMC6361815 DOI: 10.3389/fimmu.2019.00090] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/14/2019] [Indexed: 01/06/2023] Open
Abstract
Glycosphingolipids (GSLs) exhibit a variety of functions in cellular differentiation and interaction. Also, they are known to play a role as receptors in pathogen invasion. A less well-explored feature is the role of GSLs in immune cell function which is the subject of this review article. Here we summarize knowledge on GSL expression patterns in different immune cells. We review the changes in GSL expression during immune cell development and differentiation, maturation, and activation. Furthermore, we review how immune cell GSLs impact membrane organization, molecular signaling, and trans-interactions in cellular cross-talk. Another aspect covered is the role of GSLs as targets of antibody-based immunity in cancer. We expect that recent advances in analytical and genome editing technologies will help in the coming years to further our knowledge on the role of GSLs as modulators of immune cell function.
Collapse
Affiliation(s)
- Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Antonius A de Waard
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
31
|
Ouyang G, Xu Z, Jiang D, Zhu H, Wang Y, Wu W, Sun Y, Sheng L, Xu K, Lou Y, Mu Q, Zhang Y, Wu N, Cheng J, Duan S. Clinically useful flow cytometry approach to identify immunophenotype in acute leukemia. J Int Med Res 2019; 47:1483-1492. [PMID: 30614357 PMCID: PMC6460589 DOI: 10.1177/0300060518819637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES Acute leukemia (AL) is a highly heterogeneous malignant disease caused by hematopoietic cell abnormalities. Our study investigated the potential for immunophenotyping of leukemic cells via flow cytometry and the clinical usefulness of this approach in treatment of AL. METHODS Bone marrow (BM) specimens were collected to detect antigen expression on hematopoietic cells in pre-treatment samples from patients with AL. In addition, fraction survival curves were calculated using the Kaplan-Meier method to explore the effect of markers on prognosis in AL. RESULTS Expression levels of immunophenotypic markers in patients with acute lymphoblastic leukemia (ALL) were significantly different from those in patients with acute myeloid leukemia (AML). In addition, there was a potential association between the surface marker, cluster of differentiation 2 (CD2), and fraction survival in AML. However, no similar result was found in ALL. Moreover, genetic tests showed greater positive variation of the break point cluster-Abelson tyrosine kinase ( BCR-ABL) fusion gene in samples from patients with ALL than in samples from patients with AML. CONCLUSIONS We have shown a rapid and effective flow cytometry method that enables the identification of immunophenotype in AL. Moreover, CD2 may constitute a predictive marker for prognosis in patients with AML.
Collapse
Affiliation(s)
- Guifang Ouyang
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China.,*These authors contributed equally to this work
| | - Zhijuan Xu
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China.,*These authors contributed equally to this work
| | - Danjie Jiang
- 2 School of Medicine, Ningbo University, Ningbo, Zhejiang, China.,*These authors contributed equally to this work
| | - Huiling Zhu
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Yi Wang
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Wenmiao Wu
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Yongcheng Sun
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Lixia Sheng
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Kaihong Xu
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Yanru Lou
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Qitian Mu
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Yi Zhang
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Ningning Wu
- 1 Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Jia Cheng
- 2 School of Medicine, Ningbo University, Ningbo, Zhejiang, China.,a Present address: Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Shiwei Duan
- 2 School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
32
|
Brooimans RA, van der Velden VHJ, Boeckx N, Slomp J, Preijers F, Te Marvelde JG, Van NM, Heijs A, Huys E, van der Holt B, de Greef GE, Kelder A, Schuurhuis GJ. Immunophenotypic measurable residual disease (MRD) in acute myeloid leukemia: Is multicentric MRD assessment feasible? Leuk Res 2018; 76:39-47. [PMID: 30553189 DOI: 10.1016/j.leukres.2018.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/01/2018] [Accepted: 11/26/2018] [Indexed: 10/27/2022]
Abstract
Flow-cytometric detection of now termed measurable residual disease (MRD) in acute myeloid leukemia (AML) has proven to have an independent prognostic impact. In a previous multicenter study we developed protocols to accurately define leukemia-associated immunophenotypes (LAIPs) at diagnosis. It has, however, not been demonstrated whether the use of the defined LAIPs in the same multicenter setting results in a high concordance between centers in MRD assessment. In the present paper we evaluated whether interpretation of list-mode data (LMD) files, obtained from MRD assessment of previously determined LAIPs during and after treatment, could reliably be performed in a multicenter setting. The percentage of MRD positive cells was simultaneously determined in totally 173 LMD files from 77 AML patients by six participating centers. The quantitative concordance between the six participating centers was meanly 84%, with slight variation of 75%-89%. In addition our data showed that the type and number of LAIPs were of influence on the performance outcome. The highest concordance was observed for LAIPs with cross-lineage expression, followed by LAIPs with an asynchronous antigen expression. Our results imply that immunophenotypic MRD assessment in AML will only be feasible when fully standardized methods are used for reliable multicenter assessment.
Collapse
Affiliation(s)
- Rik A Brooimans
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Laboratory of Clinical and Tumor Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Vincent H J van der Velden
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nancy Boeckx
- Laboratory of Experimental Transplantation, University of Leuven, Leuven, Belgium
| | - Jennita Slomp
- Department of Clinical Chemistry, Medisch Spectrum Twente/Medlon, Enschede, The Netherlands
| | - Frank Preijers
- Department of Laboratory Medicine-Laboratory for Hematology, Radboud UMC, Nijmegen, The Netherlands
| | - Jeroen G Te Marvelde
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ngoc M Van
- Laboratory of Clinical and Tumor Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Antoinette Heijs
- Department of Clinical Chemistry, Medisch Spectrum Twente/Medlon, Enschede, The Netherlands
| | - Erik Huys
- Department of Laboratory Medicine-Laboratory for Hematology, Radboud UMC, Nijmegen, The Netherlands
| | - Bronno van der Holt
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Georgine E de Greef
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Angele Kelder
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
33
|
Morsink LM, Walter RB, Ossenkoppele GJ. Prognostic and therapeutic role of CLEC12A in acute myeloid leukemia. Blood Rev 2018; 34:26-33. [PMID: 30401586 DOI: 10.1016/j.blre.2018.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/17/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
CLEC12A has recently been identified as an antigen, expressed on leukemic stem cells and leukemic blasts. Given the fact that this expression profile seems stable throughout diagnosis, treatment and relapse on leukemic blasts and leukemic stem cells, CLEC12A can be considered a highly potent and reliable marker for the detection of measurable residual disease and therefore applicable for risk stratification and prognostication in AML. Low CLEC12A expression on leukemic blasts seems to be independently associated with lower likelihood of achieving complete remission after 1 cycle of induction chemotherapy, shorter event free survival, as well as overall survival, indicating potential prognostic properties of CLEC12A expression itself. Lack of expression on the normal hematopoietic stem and progenitor cells, in contrast to CD123 and CD33, might result in less toxicity regarding cytopenias, making CLEC12A an interesting target for innovating immunotherapies, including monoclonal and bispecific antibodies, antibody-drug conjugates and CAR-T cells therapy.
Collapse
Affiliation(s)
- Linde M Morsink
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands.
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Gert J Ossenkoppele
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
34
|
Gaipa G, Buracchi C, Biondi A. Flow cytometry for minimal residual disease testing in acute leukemia: opportunities and challenges. Expert Rev Mol Diagn 2018; 18:775-787. [DOI: 10.1080/14737159.2018.1504680] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Giuseppe Gaipa
- Department of Pediatrics, University of Milano-Bicocca, Fondazione Tettamanti - Centro Ricerca M.Tettamanti, Monza, Italy
| | - Chiara Buracchi
- Department of Pediatrics, University of Milano-Bicocca, Fondazione Tettamanti - Centro Ricerca M.Tettamanti, Monza, Italy
| | - A Biondi
- Department of Pediatrics, University of Milano-Bicocca, Fondazione Tettamanti - Centro Ricerca M.Tettamanti, Monza, Italy
- Fondazione MBBM/Ospedale San Gerardo - Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Assessment of measurable residual disease (MRD) after treatment can identify patients with acute myeloid leukemia (AML) that are at high risk of poor outcomes. However, there is no consensus yet regarding a standardized approach to measuring MRD that is most clinically meaningful. We review multiparameter flow cytometry (MFC) and reverse transcriptase polymerase chain reaction (RT-PCR), and discuss a framework for assessing remission MRD using next-generation sequencing (NGS). RECENT FINDINGS MFC and RT-PCR may not fully capitalize on the major advances that have been made in characterizing the genetic landscape of AML, which has offered insight into the biological and clinical implications of clonal genetic architecture. NGS has increasingly been shown to provide a qualitative and quantitative assessment of MRD with significant prognostic implications. The assessment of clonal architecture by NGS may complement or extend existing approaches for MRD monitoring. Long-term serial monitoring of diagnostic, remission, and relapse samples with clinical correlation will need to be performed in order to determine the impact of various MRD patterns using this technique.
Collapse
|
36
|
Mambet C, Chivu-Economescu M, Matei L, Necula LG, Dragu DL, Bleotu C, Diaconu CC. Murine models based on acute myeloid leukemia-initiating stem cells xenografting. World J Stem Cells 2018; 10:57-65. [PMID: 29988882 PMCID: PMC6033712 DOI: 10.4252/wjsc.v10.i6.57] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/24/2018] [Accepted: 06/08/2018] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive malignant disease defined by abnormal expansion of myeloid blasts. Despite recent advances in understanding AML pathogenesis and identifying their molecular subtypes based on somatic mutations, AML is still characterized by poor outcomes, with a 5-year survival rate of only 30%-40%, the majority of the patients dying due to AML relapse. Leukemia stem cells (LSC) are considered to be at the root of chemotherapeutic resistance and AML relapse. Although numerous studies have tried to better characterize LSCs in terms of surface and molecular markers, a specific marker of LSC has not been found, and still the most universally accepted phenotypic signature remains the surface antigens CD34+CD38- that is shared with normal hematopoietic stem cells. Animal models provides the means to investigate the factors responsible for leukemic transformation, the intrinsic differences between secondary post-myeloproliferative neoplasm AML and de novo AML, especially the signaling pathways involved in inflammation and hematopoiesis. However, AML proved to be one of the hematological malignancies that is difficult to engraft even in the most immunodeficient mice strains, and numerous ongoing attempts are focused to develop "humanized mice" that can support the engraftment of LSC. This present review is aiming to introduce the field of AML pathogenesis and the concept of LSC, to present the current knowledge on leukemic blasts surface markers and recent attempts to develop best AML animal models.
Collapse
Affiliation(s)
- Cristina Mambet
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Mihaela Chivu-Economescu
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania.
| | - Lilia Matei
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Laura Georgiana Necula
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Denisa Laura Dragu
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Coralia Bleotu
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Carmen Cristina Diaconu
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| |
Collapse
|
37
|
Shahni A, Saud M, Siddiqui S, Mukry SN. Expression of aberrant antigens in hematological malignancies: A single center experience. Pak J Med Sci 2018; 34:457-462. [PMID: 29805426 PMCID: PMC5954397 DOI: 10.12669/pjms.342.13996] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background and Objective Aberrant phenotype is a phenomenon of abnormal expression or loss of expression of cell specific lineage marker not associated with specific cell type. Aberrant phenotype expression due to genetic defects may be associated with unfavorable outcome. It can be used to determine minimal residual disease status. The purpose of the study was to find out the occurrence of aberrant phenotypes in leukemia/lymphoma patients. Methods One milliliter peripheral blood or bone marrow samples were analyzed on FACS Calibur flowcytometer. The cells were lysed and stained following standard protocol. Data was acquired and analyzed by CellQuest-Pro software. The Antigenic expression was rated as positive when the percentage of positive blast cells was ≥ 20%. In that manner, aberrant phenotype was considered positive when 20% of blast cells show expression of markers. Results Of a total 145 cases analyzed, 26 were acute myeloid leukemia, 71 of acute lymphoblastic leukaemia, 48 were of Chronic Lymphoid leukemia on the basis of morphological features and confirmed by flow cytometry. Overall, 19% (28) cases showed aberrant expression of antigens. In 32% (9/28) AML patients, CD5, CD7, CD64dim, CD10, CD117, CD25 and TdT were expressed while in 25% (7/28) ALL patients CD33, CD13, HLA-DR and CD3 were detected. Among chronic leukemia, all aberrant expressions were seen in cases of B-CLL (10/28) only; with CD11c, CD3 and CD10 as the aberrantly expressed markers. Conclusion Variability in aberrant phenotype expression was observed in different types of acute and chronic leukemia patients with no prognostic implications on treatment response.
Collapse
Affiliation(s)
- Aneeta Shahni
- Aneeta Shahni, BS (Clinical Laboratory Sciences). National Institute of Blood Diseases & Bone Marrow Transplantation, ST 2/A Block 17 Gulshan-e-Iqbal KDA Scheme 24, Karachi, Pakistan
| | - Madiha Saud
- Madiha Saud, M.Sc. National Institute of Blood Diseases & Bone Marrow Transplantation, ST 2/A Block 17 Gulshan-e-Iqbal KDA Scheme 24, Karachi, Pakistan
| | - Saima Siddiqui
- Saima Siddiqui, MBBS, FCPS. National Institute of Blood Diseases & Bone Marrow Transplantation, ST 2/A Block 17 Gulshan-e-Iqbal KDA Scheme 24, Karachi, Pakistan
| | - Samina Naz Mukry
- Samina Naz Mukry, Ph.D. National Institute of Blood Diseases & Bone Marrow Transplantation, ST 2/A Block 17 Gulshan-e-Iqbal KDA Scheme 24, Karachi, Pakistan
| |
Collapse
|
38
|
Merino A, Boldú L, Ermens A. Acute myeloid leukaemia: How to combine multiple tools. Int J Lab Hematol 2018; 40 Suppl 1:109-119. [DOI: 10.1111/ijlh.12831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/23/2018] [Indexed: 10/16/2022]
Affiliation(s)
- A. Merino
- Biomedical Diagnostic Center, Hospital Clínic; University of Barcelona; Barcelona Spain
| | - L. Boldú
- Biomedical Diagnostic Center, Hospital Clínic; University of Barcelona; Barcelona Spain
| | - A. Ermens
- Laboratory for Clinical Chemistry and Hematology; Amphia Hospital; Breda The Netherlands
| |
Collapse
|
39
|
Schuurhuis GJ, Heuser M, Freeman S, Béné MC, Buccisano F, Cloos J, Grimwade D, Haferlach T, Hills RK, Hourigan CS, Jorgensen JL, Kern W, Lacombe F, Maurillo L, Preudhomme C, van der Reijden BA, Thiede C, Venditti A, Vyas P, Wood BL, Walter RB, Döhner K, Roboz GJ, Ossenkoppele GJ. Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party. Blood 2018; 131:1275-1291. [PMID: 29330221 PMCID: PMC5865231 DOI: 10.1182/blood-2017-09-801498] [Citation(s) in RCA: 792] [Impact Index Per Article: 113.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022] Open
Abstract
Measurable residual disease (MRD; previously termed minimal residual disease) is an independent, postdiagnosis, prognostic indicator in acute myeloid leukemia (AML) that is important for risk stratification and treatment planning, in conjunction with other well-established clinical, cytogenetic, and molecular data assessed at diagnosis. MRD can be evaluated using a variety of multiparameter flow cytometry and molecular protocols, but, to date, these approaches have not been qualitatively or quantitatively standardized, making their use in clinical practice challenging. The objective of this work was to identify key clinical and scientific issues in the measurement and application of MRD in AML, to achieve consensus on these issues, and to provide guidelines for the current and future use of MRD in clinical practice. The work was accomplished over 2 years, during 4 meetings by a specially designated MRD Working Party of the European LeukemiaNet. The group included 24 faculty with expertise in AML hematopathology, molecular diagnostics, clinical trials, and clinical medicine, from 19 institutions in Europe and the United States.
Collapse
Affiliation(s)
- Gerrit J Schuurhuis
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Sylvie Freeman
- Department of Clinical Immunology, Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Francesco Buccisano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pediatric Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - David Grimwade
- Division of Genetics & Molecular Medicine, King's College, London, United Kingdom
| | | | - Robert K Hills
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | | | - Jeffrey L Jorgensen
- Division of Pathology/Laboratory Medicine, Department of Hematopathology, MD Anderson Cancer Center, Houston, TX
| | | | - Francis Lacombe
- Flow Cytometry Platform, University Hospital, Bordeaux, France
| | - Luca Maurillo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Claude Preudhomme
- Center of Pathology, Laboratory of Hematology, University Hospital of Lille, Lille, France
| | - Bert A van der Reijden
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian Thiede
- Universitätsklinikum Carl Gustav Garus an der Technischen Universität Dresden, Dresden, Germany
| | - Adriano Venditti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Paresh Vyas
- Medical Research Council Molecular Haematology Unit, Oxford Centre for Haematology, University of Oxford and Oxford University Hospitals National Health Service Trust, Oxford, United Kingdom
| | - Brent L Wood
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Laboratory Medicine and
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany; and
| | - Gail J Roboz
- Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY
| | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Molecular Heterogeneity in Acute Promyelocytic Leukemia - a Single Center Experience from India. Mediterr J Hematol Infect Dis 2018; 10:e2018002. [PMID: 29326799 PMCID: PMC5760075 DOI: 10.4084/mjhid.2018.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/06/2017] [Indexed: 11/24/2022] Open
Abstract
Atypical breakpoints and variant APL cases involving alternative chromosomal aberrations are seen in a small subset of acute promyelocytic leukemia (APL) patients. Over seven different partner genes for RARA have been described. Although rare, these variants prove to be a diagnostic challenge and require a combination of advanced cytogenetic and molecular techniques for accurate characterization. Heterogeneity occurs not only at the molecular level but also at clinico-pathological level influencing treatment response and outcome. In this case series, we describe the molecular heterogeneity of APL with a focus on seven variant APL cases from a single tertiary cancer center in India over a period of two and a half years. We discuss five cases with ZBTB16-RARA fusion and two novel PML-RARA variants, including a Bcr3 variant involving fusion of PML exon4 and RARA exon3 with an additional 40 nucleotides originating from RARA intron2, another involving exon 6 of PML and exon 3 of RARA with addition of 126 nucleotides, which mapped to the central portion of RARA intron 2. To the best of our knowledge, this is the first case series of this kind from India.
Collapse
|
41
|
Lacombe F, Campos L, Allou K, Arnoulet C, Delabarthe A, Dumezy F, Feuillard J, Geneviève F, Guérin E, Guy J, Jouault H, Lepelley P, Maynadié M, Solly F, Ballon OW, Preudhomme C, Baruchel A, Dombret H, Ifrah N, Béné MC. Prognostic value of multicenter flow cytometry harmonized assessment of minimal residual disease in acute myeloblastic leukemia. Hematol Oncol 2017; 36:422-428. [DOI: 10.1002/hon.2488] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/13/2017] [Accepted: 11/06/2017] [Indexed: 01/17/2023]
Affiliation(s)
| | - Lydia Campos
- Hematology Biology; University Hospital; Saint Etienne France
| | - Kaoutar Allou
- Hematology Biology; University Hospital; Bordeaux France
| | | | | | | | | | | | | | - Julien Guy
- Hematology Biology; University Hospital; Dijon France
| | | | | | - Marc Maynadié
- Hematology Biology; University Hospital; Dijon France
| | - Françoise Solly
- Hematology Biology; University Hospital; Saint Etienne France
| | | | | | - André Baruchel
- Hematology Department; Hôpital Robert Debré; Paris France
| | - Hervé Dombret
- Hematology Department; Hôpital Saint Louis; Paris France
| | - Norbert Ifrah
- Hematology Department; University Hospital; Angers France
| | - Marie C. Béné
- Hematology Biology; University Hospital; Nantes France
| | | |
Collapse
|
42
|
Getta BM, Roshal M, Zheng J, Park JH, Stein EM, Levine R, Papadopoulos EB, Jakubowski AA, Kernan NA, Steinherz P, O'Reilly RJ, Perales MA, Giralt SA, Tallman MS, Shaffer BC. Allogeneic Hematopoietic Stem Cell Transplantation with Myeloablative Conditioning Is Associated with Favorable Outcomes in Mixed Phenotype Acute Leukemia. Biol Blood Marrow Transplant 2017; 23:1879-1886. [PMID: 28694182 PMCID: PMC5682215 DOI: 10.1016/j.bbmt.2017.06.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022]
Abstract
Mixed phenotype acute leukemia (MPAL) represents a poorly characterized group of acute leukemias that lack an accepted therapeutic approach and are typically associated with poor outcomes. We present our experience of genomic profiling, pretransplantation therapy, and transplantation outcomes for 36 well-characterized pediatric and adult patients with MPAL, defined according to the 2016 World Health Organization leukemia update. A predominance of acute lymphoid leukemia (ALL)-associated mutations and cytogenetic abnormalities was noted. Remission rates after induction appeared comparable among adults (20 of 23) and children (11 of 13) and among those who received ALL (10 of 11) or acute myeloid leukemia-type (21 of 25) induction. Adults underwent transplantation in first remission while children underwent transplantation in the setting of relapse or MLL rearrangement. The median follow-up among the 25 patients who underwent transplantation was 39.6 months and median overall survival was not reached. Relapse after transplantation was associated with MLL rearrangement (P = .022), reduced-intensity conditioning (P < .001), and higher WBC at diagnosis (P = .034). These data highlight differing therapeutic approaches between adult and pediatric MPAL and demonstrate favorable survival of adult MPAL patients consolidated with allogeneic hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Bartlomiej M Getta
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Junting Zheng
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jae H Park
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Eytan M Stein
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Ross Levine
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Esperanza B Papadopoulos
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Ann A Jakubowski
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Nancy A Kernan
- Weill Cornell Medical College, New York, New York; Pediatric Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter Steinherz
- Weill Cornell Medical College, New York, New York; Leukemia Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J O'Reilly
- Weill Cornell Medical College, New York, New York; Pediatric Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Sergio A Giralt
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Martin S Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Brian C Shaffer
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York.
| |
Collapse
|
43
|
Schneider C, Bayerl M, Boyer C, Desai R, Claxton D, Van de Louw A. Increased CD13 Expression in Acute Myeloid Leukemia-associated Early Acute Hypoxic Respiratory Failure. Am J Respir Crit Care Med 2017; 196:1077-1080. [PMID: 28323433 DOI: 10.1164/rccm.201701-0080le] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Coursen Schneider
- 1 Penn State Milton S. Hershey Medical Center and College of Medicine Hershey, Pennsylvania
| | - Michael Bayerl
- 1 Penn State Milton S. Hershey Medical Center and College of Medicine Hershey, Pennsylvania
| | - Cinda Boyer
- 1 Penn State Milton S. Hershey Medical Center and College of Medicine Hershey, Pennsylvania
| | - Ruchi Desai
- 1 Penn State Milton S. Hershey Medical Center and College of Medicine Hershey, Pennsylvania
| | - David Claxton
- 1 Penn State Milton S. Hershey Medical Center and College of Medicine Hershey, Pennsylvania
| | - Andry Van de Louw
- 1 Penn State Milton S. Hershey Medical Center and College of Medicine Hershey, Pennsylvania
| |
Collapse
|
44
|
Laing AA, Harrison CJ, Gibson BE, Keeshan K. Unlocking the potential of anti-CD33 therapy in adult and childhood acute myeloid leukemia. Exp Hematol 2017; 54:40-50. [DOI: 10.1016/j.exphem.2017.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 10/19/2022]
|
45
|
Voigt AP, Brodersen LE, Alonzo TA, Gerbing RB, Menssen AJ, Wilson ER, Kahwash S, Raimondi SC, Hirsch BA, Gamis AS, Meshinchi S, Wells DA, Loken MR. Phenotype in combination with genotype improves outcome prediction in acute myeloid leukemia: a report from Children's Oncology Group protocol AAML0531. Haematologica 2017; 102:2058-2068. [PMID: 28883080 PMCID: PMC5709105 DOI: 10.3324/haematol.2017.169029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/06/2017] [Indexed: 12/17/2022] Open
Abstract
Diagnostic biomarkers can be used to determine relapse risk in acute myeloid leukemia, and certain genetic aberrancies have prognostic relevance. A diagnostic immunophenotypic expression profile, which quantifies the amounts of distinct gene products, not just their presence or absence, was established in order to improve outcome prediction for patients with acute myeloid leukemia. The immunophenotypic expression profile, which defines each patient’s leukemia as a location in 15-dimensional space, was generated for 769 patients enrolled in the Children’s Oncology Group AAML0531 protocol. Unsupervised hierarchical clustering grouped patients with similar immunophenotypic expression profiles into eleven patient cohorts, demonstrating high associations among phenotype, genotype, morphology, and outcome. Of 95 patients with inv(16), 79% segregated in Cluster A. Of 109 patients with t(8;21), 92% segregated in Clusters A and B. Of 152 patients with 11q23 alterations, 78% segregated in Clusters D, E, F, G, or H. For both inv(16) and 11q23 abnormalities, differential phenotypic expression identified patient groups with different survival characteristics (P<0.05). Clinical outcome analysis revealed that Cluster B (predominantly t(8;21)) was associated with favorable outcome (P<0.001) and Clusters E, G, H, and K were associated with adverse outcomes (P<0.05). Multivariable regression analysis revealed that Clusters E, G, H, and K were independently associated with worse survival (P range <0.001 to 0.008). The Children’s Oncology Group AAML0531 trial: clinicaltrials.gov Identifier: 00372593.
Collapse
Affiliation(s)
| | | | - Todd A Alonzo
- Children's Oncology Group, Monrovia, CA, USA.,University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | - Betsy A Hirsch
- University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Alan S Gamis
- Children's Mercy Hospitals & Clinics, Kansas City, MO, USA
| | - Soheil Meshinchi
- Children's Oncology Group, Monrovia, CA, USA.,Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | |
Collapse
|
46
|
Luskin MR, Stone RM. Can Minimal Residual Disease Determination in Acute Myeloid Leukemia Be Used in Clinical Practice? J Oncol Pract 2017; 13:471-480. [DOI: 10.1200/jop.2017.021675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In acute myeloid leukemia (AML) that is in complete remission, minimal residual disease (MRD) is presumed to be present, though not morphologically evident. Advances in diagnostics now permit the detection and quantification of MRD in AML by several techniques. The level of MRD after induction and consolidation therapy correlates with disease sensitivity to chemotherapy and has greater power to predict long-term survival than patient and disease characteristics that are available at diagnosis, including genetic information. A unique advantage of MRD is that it is an integrated measure of the impact and interaction of genetics, epigenetics, host immune milieu, bone marrow environment, and drug sensitivity on disease response to treatment. Here, we review the main techniques for MRD assessment in AML, including polymerase chain reaction, multiparameter flow cytometry, and next-generation sequencing, with a focus on method-specific and general limitations to the optimal employment of MRD techniques for the determination of AML prognosis. We also review the data that establish the prognostic and predictive value of MRD assessment in AML. Finally, we provide recommendations for the use of MRD in the care of patients with AML in clinical practice today, including whether it should influence treatment decisions.
Collapse
Affiliation(s)
- Marlise R. Luskin
- Dana-Farber Cancer Institute; and Harvard Medical School, Boston, MA
| | - Richard M. Stone
- Dana-Farber Cancer Institute; and Harvard Medical School, Boston, MA
| |
Collapse
|
47
|
Ossenkoppele G, Schuurhuis GJ. MRD in AML: does it already guide therapy decision-making? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:356-365. [PMID: 27913502 PMCID: PMC6142473 DOI: 10.1182/asheducation-2016.1.356] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Prognostic factors determined at diagnosis are predictive for outcome whereas achievement of morphological complete remission (CR) is still an important end point during treatment. Residual disease after therapy may reflect the sum of all diagnosis and postdiagnosis resistance mechanisms/factors; its measurement could hypothetically be very instrumental for guiding treatment. The possibility of defining residual disease (minimal residual disease [MRD]) far below the level of 5% blast cells is changing the landscape of risk classification. In this manuscript, the various methods, all different in sensitivity, specificity, and phase of development, to assess MRD are discussed. Currently, the 2 methods mostly used are flow cytometry-based immune MRD (multiparameter flow cytometry [MPFC]) and molecular MRD assessed by real-time quantitative polymerase chain reaction. Both have advantages and disadvantages that are summarized in detail. Many studies in children as well as adults already demonstrated that MRD detection by MPFC or molecular MRD provides strong prognostic information in acute myeloid leukemia (AML) after both induction and consolidation. These studies are summarized in this review. The general conclusion of this review is that a better definition of disease burden than morphological CR is now emerging. MRD assessed by flow or molecular techniques should become standard in every clinical trial in AML. Harmonization of antibody panels, introduction of single-cell tube systems (for determination of residual leukemic stem cells), and standardized analytical programs will pave the way for individual risk assessment and become a surrogate end point for survival in studies investigating new drugs, hopefully resulting in faster drug approval in AML.
Collapse
Affiliation(s)
- Gert Ossenkoppele
- Department of Hematology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Gerrit Jan Schuurhuis
- Department of Hematology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Minimal Residual Disease in Acute Myeloid Leukemia of Adults: Determination, Prognostic Impact and Clinical Applications. Mediterr J Hematol Infect Dis 2016; 8:e2016052. [PMID: 27872732 PMCID: PMC5111512 DOI: 10.4084/mjhid.2016.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 09/12/2016] [Indexed: 02/06/2023] Open
Abstract
Pretreatment assessment of cytogenetic/genetic signature of acute myeloid leukemia (AML) has been consistently shown to play a major prognostic role but also to fail at predicting outcome on individual basis, even in low-risk AML. Therefore, we are in need of further accurate methods to refine the patients’ risk allocation process, distinguishing more adequately those who are likely to recur from those who are not. In this view, there is now evidence that the submicroscopic amounts of leukemic cells (called minimal residual disease, MRD), measured during the course of treatment, indicate the quality of response to therapy. Therefore, MRD might serve as an independent, additional biomarker to help to identify patients at higher risk of relapse. Detection of MRD requires the use of highly sensitive ancillary techniques, such as polymerase chain reaction (PCR) and multiparametric flow cytometry(MPFC). In the present manuscript, we will review the current approaches to investigate MRD and its clinical applications in AML management.
Collapse
|
49
|
Eissa DS, Kandeel EZ, Ghareeb M. Human myeloid inhibitory C-lectin: a highly specific and stable acute myeloid leukemia marker. Hematol Oncol 2016; 35:814-820. [PMID: 27734526 DOI: 10.1002/hon.2352] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/01/2016] [Accepted: 08/10/2016] [Indexed: 12/24/2022]
Abstract
The prognosis of acute myeloid leukemia (AML) is poor because of relapses occurring on conventional chemotherapy. The distinction between leukemic and normal stem cells relies on the expression of antigen combinations defining leukemia-associated immunophenotypes (LAIPs), which are absent or extremely infrequent in normal bone marrow. However, LAIPs are very different from patient to patient and are not necessarily stable over the course of the disease. Accordingly, we addressed the applicability of human myeloid inhibitory C-lectin (hMICL) by flow cytometry as a specific leukemic myeloid stem cell marker for the diagnosis of AML in CD34+ and CD34- cases and evaluated the stability of hMICL during the course of the disease. hMICL expression was assessed in 78 bone marrow aspirate specimens obtained from AML patients at diagnosis (n = 40), complete remission (CR) (n = 28), and relapse (n = 10). AML patients at diagnosis were compared to 20 newly diagnosed acute lymphoblastic leukemia (ALL) patients and 20 healthy controls. hMICL was reevaluated in CR and relapse specimens. hMICL was expressed in 100% AML patients at diagnosis (mean ± standard deviation [SD], 60.3 ± 19.9%), both CD34+ and CD34- , but not in ALL (mean ± SD, 3.3 ± 1.9%) or healthy controls (mean ± SD, 3.4 ± 2.6%) (P < .001). hMICL median fluorescence intensity ratio was higher in AML (mean ± SD, 15.9 ± 11.7) compared to ALL (mean ± SD, 4.5 ± 1.4) and healthy controls (mean ± SD, 4.4 ± 1.6) (P < .001). hMICL was expressed in all studied AML morphologic subtypes. Preserved stable expression of hMICL was found in CR and relapse specimens with no antigen loss. hMICL is a robust pan-AML-associated antigen with excellent diagnostic impact, extreme specificity to AML blasts, and stability throughout the course of the disease. hMICL could be incorporated into the routine flow cytometry setting within the initial diagnostic work-up and follow-up of AML.
Collapse
Affiliation(s)
- Deena Samir Eissa
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman Zaghloul Kandeel
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Ghareeb
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
50
|
Ossenkoppele GJ, Janssen JJWM, van de Loosdrecht AA. Risk factors for relapse after allogeneic transplantation in acute myeloid leukemia. Haematologica 2016; 101:20-5. [PMID: 26721801 DOI: 10.3324/haematol.2015.139105] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acute myeloid leukemia is a clonal neoplasm derived from myeloid progenitor cells with a varying outcome. The initial goal of treatment is the achievement of complete remission, defined for over 40 years by morphology. However, without additional post-remission treatment the majority of patients relapse. In many cases of acute myeloid leukemia, allogeneic stem cell transplantation offers the best prospects of cure. In 2013, 5608 stem cell transplantations in acute myeloid leukemia were performed in Europe (5228 allogeneic and 380 autologous stem cell transplantations). Most stem cell transplantations are performed in first complete remission. However, despite a considerable reduction in the chance of relapse, in most studies, overall survival benefit of allogeneic stem cell transplantation is modest due to substantial non-relapse mortality. Here we discuss the many factors related to the risk of relapse after allogeneic stem cell transplantation.
Collapse
|