1
|
Firrera L, Podvin B, Herlem J, Magierowicz M, Willaume A, Thibaud V, Charpentier A. A New Diagnostic Approach for Myelodysplastic Neoplasms Using a Combination of Scores Based on Flow Cytometry and Automated Hematology Sysmex XN Analyzers. Int J Lab Hematol 2025; 47:236-245. [PMID: 39658949 PMCID: PMC11885685 DOI: 10.1111/ijlh.14404] [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: 07/10/2024] [Revised: 09/27/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024]
Abstract
INTRODUCTION The first-step in diagnosis of myelodysplastic neoplasms (MDS) is essentially based on bone marrow cytomorphology. However, cytomorphology of MDS is often a difficult exercise, subject to inter-operator variability. Our study aims to evaluate whether the combination of two dysplasia scores, the extended Ogata score and the MDS-CBC score, could improve the screening of MDS patients among patients with chronic cytopenia. METHODS Extended Ogata score and MDS-CBC score have been measured on a retrospective cohort of 63 patients with a clinical suspicion of MDS based on the presence of cytopenia. Among these patients, 33 patients were diagnosed as MDS (MDS group) and 30 patients were diagnosed with another cause of cytopenia (non-MDS cytopenic control group). RESULTS Our results show excellent performance of the combined scores in predicting MDS when the two scores are concordant: positive predictive value (PPV) = 96% and negative predictive value (NPV) = 92%. In comparison, in the same cohort, extended Ogata score alone showed a PPV = 90% and NPV = 79%, MDS-CBC score alone showed a PPV = 85% and NPV = 86%. CONCLUSION For the first time, our results show that the combination of these two dysplasia scores constitutes a useful and rapid tool for the assessment of dysplasia associated with MDS. In the MDS diagnostic process, the use of combined scores could constitute a valuable tool to enable early strong prediction of MDS in cytopenic patients and to target patients who initially require additional genetic assays.
Collapse
Affiliation(s)
- Ludovic Firrera
- Laboratory of HematologyHospital Group of Lille Catholic UniversityLilleFrance
| | - Benjamin Podvin
- Laboratory of HematologyHospital Group of Lille Catholic UniversityLilleFrance
- Laboratory of HematologyCHU LilleLilleFrance
| | - Julien Herlem
- Laboratory of HematologyHospital Group of Lille Catholic UniversityLilleFrance
| | - Marion Magierowicz
- Laboratory of HematologyHospital Group of Lille Catholic UniversityLilleFrance
| | - Alexandre Willaume
- Hematology DepartmentHospital Group of Lille Catholic UniversityLilleFrance
| | - Vincent Thibaud
- Hematology DepartmentHospital Group of Lille Catholic UniversityLilleFrance
| | - Agnès Charpentier
- Laboratory of HematologyHospital Group of Lille Catholic UniversityLilleFrance
| |
Collapse
|
2
|
Ecker V, Müller ML, Kern W. Flow Cytometric Bone Marrow Evaluation in Suspected Myelodysplastic Neoplasms. Curr Protoc 2025; 5:e70137. [PMID: 40256960 DOI: 10.1002/cpz1.70137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Myelodysplastic neoplasms (MDS) are acquired heterogeneous clonal hematopoietic stem cell neoplasms, clinically characterized by progressively ineffective hematopoiesis and an increased risk of acute myeloid leukemia. MDS are accompanied by an inflammatory microenvironment and genome instability. Signs of dysplasia can occur in the erythroid, myeloid, monocytic, and megakaryocytic cell lineages and result in anemia, neutropenia, and thrombocytopenia. Multi-parameter flow cytometry can be used to detect aberrant antigen expression patterns typical of MDS, which correlate with cytomorphologically identified dysplasias and provide important information for diagnosis and prognosis. Characteristic findings include an increase in myeloid progenitor cells; aberrant myeloid and erythroid maturation; aberrant marker expression on progenitor cells, granulocytes, and monocytes, which corresponds to lineage infidelity, under-/overexpression, or asynchronous expression; and an increase in monocytes and progenitor cells in chronic myelomonocytic leukemia. The latter represents an independent disease entity with a similar phenotype. In addition, flow cytometry can rule out other causes of cytopenia, such as lymphoma, acute leukemias, paroxysmal nocturnal hemoglobinuria, or systemic mastocytosis with associated hematologic neoplasm. To analyze those features, the European LeukemiaNet recommends a set of markers together with important technical aspects. At least three distinct aberrations in at least two lineages are associated with a high likelihood of MDS. © 2025 Wiley Periodicals LLC. Basic Protocol: Flow cytometric bone marrow evaluation in suspected myelodysplastic neoplasms.
Collapse
|
3
|
Valerio Ikoma-Colturato MR, Furtado FM, de Oliveira E, Gevert F, Mendonça R. How I Investigate Measurable Residual Disease in B-Cell Precursor Acute Lymphoblastic Leukemia After Therapy With Bi-Specific Monoclonal Antibodies and 19CAR-T Cells. Int J Lab Hematol 2025. [PMID: 40007453 DOI: 10.1111/ijlh.14448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/12/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION Measurable residual disease (MRD) in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) following anti-CD19 targeted therapies requires specific strategies to identify residual blast cells due to loss or reduced CD19 expression that makes it inconsistent as a primitive marker for B-cell gating. OBJECTIVE Due to the increased access of BCP-ALL patients to therapies with CD3/CD19 bispecific T-cell engagers (BiTe) and CD19-targeted chimeric antigen receptor T-Cell (CAR-T), it is essential that flow cytometry laboratories are prepared to evaluate therapeutic responses. MATERIAL AND METHODS Here, validated strategies for MRD detection in the context of anti-CD19 therapies are described, accessible to flow cytometry laboratories according to their different facilities. The paper includes an 8-color flow cytometry (FC) strategy for BCP-ALL MRD based on alternative gating without the use of additional markers (Euroflow protocol), as well as other strategies using alternative markers to CD19, comprising 2 protocols using 8 colors, one using 10 colors and another 14 colors/15 markers. CONCLUSION Different strategies are needed to detect MRD without using CD19 for B-cell population gating after CD19-targeted therapies. However, it is essential that validated protocols are used according to the available resources to ensure reliable results for clinical decision-making.
Collapse
Affiliation(s)
| | - Felipe Magalhães Furtado
- Hospital da Criança de Brasília José Alencar, Brasília, Brazil
- Sabin Medicina Diagnóstica, Brasília, Brazil
| | - Elen de Oliveira
- Instituto de Puericultura e Pediatria e Martagão Gesteira (IPPMG/UFRJ), Rio de Janeiro, Brazil
| | - Fabiola Gevert
- Hospital Pequeno Príncipe, Curitiba, Brazil
- Hospital Erasto Gaertner, Curitiba, Brazil
| | | |
Collapse
|
4
|
Oster HS, Van de Loosdrecht AA, Mittelman M. Diagnosis of myelodysplastic syndromes: the classic and the novel. Haematologica 2025; 110:300-311. [PMID: 39445407 PMCID: PMC11788627 DOI: 10.3324/haematol.2023.284937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
The myelodysplastic syndromes (MDS) are a heterogenous group of clonal bone marrow (BM) stem cell myeloid neoplasms, characterized by BM dysplasia, macrocytic anemia or cytopenia with a tendency for leukemic transformation. The suspicion of MDS is raised by a typical but not specific clinical picture and routine laboratory findings, but the gold standard for the diagnosis of MDS is still BM examination with the presence of uni-or multi-lineage dysplasia and blast percentage, together with exclusion of other reasons. Cytogenetics is also a part of the diagnostic process. Flow cytometry and genetics are helpful but are not always mandatory for the diagnosis of MDS. This review summarizes the current steps in the diagnostic approach for a patient suspected of having MDS. We also describe new concepts that use non-invasive diagnostic technologies, especially digital methods as well as peripheral blood genetics. The hope is that one day these will mature, be introduced into clinical practice, and perhaps in many cases even replace the invasive BM biopsy.
Collapse
Affiliation(s)
- Howard S Oster
- Department of Medicine, Tel Aviv Sourasky Medical Center, Tel Aviv University School of Medicine
| | - Arjan A Van de Loosdrecht
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam
| | - Moshe Mittelman
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv University School of Medicine.
| |
Collapse
|
5
|
Wang D, Fang H, Ok CY, Jorgensen JL, Medeiros LJ, Wang W, Wang SA. Advancing Diagnostic Accuracy and Quality of Patient Care Through the Implementation of a Flow Cytometry Quality Assurance Program. Arch Pathol Lab Med 2025; 149:e26-e30. [PMID: 38871355 DOI: 10.5858/arpa.2024-0020-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 06/15/2024]
Abstract
CONTEXT.— Flow cytometry immunophenotypic analysis plays an important role in the diagnosis, classification, and disease monitoring of hematologic neoplasms. The interpretation of flow cytometry testing can be challenging. OBJECTIVE.— To explore ways to improve diagnostic accuracy and in turn enhance the quality of patient care. DESIGN.— A flow cytometry quality assurance (QA) program was developed. Cases from various complex flow cytometry panels were randomly selected and cross-reviewed. The outcomes of the QA review were categorized into 3 groups: complete agreement, minor discrepancy, and major discrepancy. Each discrepancy underwent a process of documentation, discussion, and resolution. Here we summarize our 3 years of experience with this program. RESULTS.— In total, 6166 cases were evaluated; 6028 cases (97.7%) showed complete concordance, 120 cases (2.0%) showed minor discrepancies, and 18 cases (0.3%) showed major discrepancies. Among the top 5 panels evaluated, the panel evaluating mature T-cell abnormalities showed the highest rate of discrepancy, whereas the panel for evaluation of myelodysplastic syndromes showed the lowest discrepancy rate. When analyzing the trends of concordance and discrepancy over time, we observed a statistically significant decrease in discrepancy rate over time, from 4% at the beginning of the 6-month period to 1.5% in the final 6-month period. CONCLUSIONS.— The overall concordance rate was 97.7%. The remaining 2.3% of cases showed discrepancies that required a correction, underscoring the value and necessity of having a QA program. The overall discrepancy rates exhibited a gradual decline over time, indicative of the positive impact of the QA program on enhancing diagnostic competency and accuracy over time.
Collapse
Affiliation(s)
- Dylan Wang
- From the Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Hong Fang
- From the Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Chi Young Ok
- From the Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jeffrey L Jorgensen
- From the Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston
| | - L Jeffrey Medeiros
- From the Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Wei Wang
- From the Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Sa A Wang
- From the Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
6
|
Cai Q, Lan H, Yi D, Xian B, Zidan L, Li J, Liao Z. Flow cytometry in acute myeloid leukemia and detection of minimal residual disease. Clin Chim Acta 2025; 564:119945. [PMID: 39209245 DOI: 10.1016/j.cca.2024.119945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Acute myeloid leukemia (AML) is a common type of acute leukemia (AL), belonging to malignant tumors of the hematopoietic system with the characteristics of rapid disease development, control with extreme difficulties, easy recurrence, poor prognosis, and incidence rate increasing with age. The traditionally diagnostic standard of French American British (FAB), being based on the morphological examination with high human subjectivity, can no longer meet the demand of clinical diagnosis and treatment of AML. Requirements of objective accuracy and low-dose sample, have become the indispensable method for AML diagnosis and monitoring prognosis. Flow cytometry is a modern technology that can quickly and accurately detect the series, antigen distribution, differentiation stage of AML cells, minimal residual lesions after AML therapy, so as to provide the great significance in guiding clinical diagnosis, hierarchical treatment, and prognosis judgement. This article will systematically elaborate on the application of flow cytometry in the diagnosis and classification of AML, and the detection of minimal residual lesions, thereby providing reference significance for dynamic monitoring and prognostic observation of AML with different immune subtypes of FAB.
Collapse
Affiliation(s)
- Qihui Cai
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Haiqiang Lan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Deng Yi
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Bojun Xian
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Luo Zidan
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Jianqiao Li
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Zhaohong Liao
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
7
|
Veenbergen S, Jugooa R, Te Marvelde J, de Vries ACH, van der Velden VHJ. Normal and malignant cells are homogeneously distributed in the bone marrow of children. Leuk Lymphoma 2024; 65:1186-1189. [PMID: 38587123 DOI: 10.1080/10428194.2024.2337782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Affiliation(s)
- Sharon Veenbergen
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Romana Jugooa
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeroen Te Marvelde
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrica C H de Vries
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent H J van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
8
|
Tarfi S, Kern W, Goulas E, Selimoglu-Buet D, Wagner-Ballon O. Technical, gating and interpretation recommendations for the partitioning of circulating monocyte subsets assessed by flow cytometry. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:203-215. [PMID: 38656036 DOI: 10.1002/cyto.b.22176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/24/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
The monocyte subset partitioning by flow cytometry, known as "monocyte assay," is now integrated into the new classifications as a supporting criterion for CMML diagnosis, if a relative accumulation of classical monocytes above 94% of total circulating monocytes is observed. Here we provide clinical flow cytometry laboratories with technical support adapted for the most commonly used cytometers. Step-by-step explanations of the gating strategy developed on whole peripheral blood are presented while underlining the most common difficulties. In a second part, interpretation recommendations of circulating monocyte partitioning from the dedicated French working group "CytHem-LMMC" are shared as well as the main pitfalls, including false positive and false negative cases. The particular flow-defined inflammatory profile is described and the usefulness of the nonclassical monocyte specific marker, namely slan, highlighted. Examples of reporting to the physician with frequent situations encountered when using the monocyte assay are also presented.
Collapse
Affiliation(s)
- Sihem Tarfi
- Département d'Hématologie et Immunologie Biologiques, AP-HP, Hôpital Henri Mondor, Créteil, France
| | - Wolfgang Kern
- MLL Munich Leukemia Laboratory, GmbH, Munich, Germany
| | - Elodie Goulas
- Département d'Hématologie et Immunologie Biologiques, AP-HP, Hôpital Henri Mondor, Créteil, France
| | - Dorothée Selimoglu-Buet
- INSERM Unité Mixte de Recherche (UMR) 1287, Faculté de Médecine, Université Paris-Sud, Villejuif, France
| | - Orianne Wagner-Ballon
- Département d'Hématologie et Immunologie Biologiques, AP-HP, Hôpital Henri Mondor, Créteil, France
- INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| |
Collapse
|
9
|
Tentori CA, Zhao LP, Tinterri B, Strange KE, Zoldan K, Dimopoulos K, Feng X, Riva E, Lim B, Simoni Y, Murthy V, Hayes MJ, Poloni A, Padron E, Cardoso BA, Cross M, Winter S, Santaolalla A, Patel BA, Groarke EM, Wiseman DH, Jones K, Jamieson L, Manogaran C, Daver N, Gallur L, Ingram W, Ferrell PB, Sockel K, Dulphy N, Chapuis N, Kubasch AS, Olsnes AM, Kulasekararaj A, De Lavellade H, Kern W, Van Hemelrijck M, Bonnet D, Westers TM, Freeman S, Oelschlaegel U, Valcarcel D, Raddi MG, Grønbæk K, Fontenay M, Loghavi S, Santini V, Almeida AM, Irish JM, Sallman DA, Young NS, van de Loosdrecht AA, Adès L, Della Porta MG, Cargo C, Platzbecker U, Kordasti S. Immune-monitoring of myelodysplastic neoplasms: Recommendations from the i4MDS consortium. Hemasphere 2024; 8:e64. [PMID: 38756352 PMCID: PMC11096644 DOI: 10.1002/hem3.64] [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: 01/31/2024] [Accepted: 03/03/2024] [Indexed: 05/18/2024] Open
Abstract
Advancements in comprehending myelodysplastic neoplasms (MDS) have unfolded significantly in recent years, elucidating a myriad of cellular and molecular underpinnings integral to disease progression. While molecular inclusions into prognostic models have substantively advanced risk stratification, recent revelations have emphasized the pivotal role of immune dysregulation within the bone marrow milieu during MDS evolution. Nonetheless, immunotherapy for MDS has not experienced breakthroughs seen in other malignancies, partly attributable to the absence of an immune classification that could stratify patients toward optimally targeted immunotherapeutic approaches. A pivotal obstacle to establishing "immune classes" among MDS patients is the absence of validated accepted immune panels suitable for routine application in clinical laboratories. In response, we formed International Integrative Innovative Immunology for MDS (i4MDS), a consortium of multidisciplinary experts, and created the following recommendations for standardized methodologies to monitor immune responses in MDS. A central goal of i4MDS is the development of an immune score that could be incorporated into current clinical risk stratification models. This position paper first consolidates current knowledge on MDS immunology. Subsequently, in collaboration with clinical and laboratory specialists, we introduce flow cytometry panels and cytokine assays, meticulously devised for clinical laboratories, aiming to monitor the immune status of MDS patients, evaluating both immune fitness and identifying potential immune "risk factors." By amalgamating this immunological characterization data and molecular data, we aim to enhance patient stratification, identify predictive markers for treatment responsiveness, and accelerate the development of systems immunology tools and innovative immunotherapies.
Collapse
Affiliation(s)
- Cristina A. Tentori
- Humanitas Clinical and Research Center–IRCCS & Department of Biomedical SciencesHumanitas UniversityMilanItaly
- Comprehensive Cancer Centre, King's CollegeLondonUK
| | - Lin P. Zhao
- Hématologie seniorsHôpital Saint‐Louis, Assistance Publique des Hôpitaux de Paris (APHP)ParisFrance
- INSERM UMR_S1160, Institut de Recherche Saint LouisUniversité Paris CitéParisFrance
| | - Benedetta Tinterri
- Humanitas Clinical and Research Center–IRCCS & Department of Biomedical SciencesHumanitas UniversityMilanItaly
| | - Kathryn E. Strange
- Comprehensive Cancer Centre, King's CollegeLondonUK
- Research Group of Molecular ImmunologyFrancis Crick InstituteLondonUK
| | - Katharina Zoldan
- Department of Medicine 1, Haematology, Cellular Therapy, Hemostaseology and Infectious DiseasesUniversity Medical Center LeipzigLeipzigGermany
| | - Konstantinos Dimopoulos
- Department of Clinical BiochemistryBispebjerg and Frederiksberg HospitalCopenhagenDenmark
- Department of Pathology, RigshospitaletCopenhagen University HospitalCopenhagenDenmark
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung and Blood InstituteBethesdaMarylandUSA
| | - Elena Riva
- Humanitas Clinical and Research Center–IRCCS & Department of Biomedical SciencesHumanitas UniversityMilanItaly
| | | | - Yannick Simoni
- Université Paris Cité, CNRS, INSERM, Institut CochinParisFrance
| | - Vidhya Murthy
- Centre for Clinical Haematology, University Hospitals of BirminghamBirminghamUK
| | - Madeline J. Hayes
- Cell & Developmental BiologyVanderbilt University School of MedicineNashvilleTennesseeUSA
- Pathology, Microbiology and Immunology, Vanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt‐Ingram Cancer Center, Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Antonella Poloni
- Department of Clinical and Molecular SciencesUniversità Politecnica delle MarcheAnconaItaly
| | - Eric Padron
- Moffitt Cancer Center, Malignant Hematology DepartmentTampaUSA
| | - Bruno A. Cardoso
- Universidade Católica PortuguesaFaculdade de MedicinaPortugal
- Universidade Católica Portuguesa, Centro de Investigação Interdisciplinar em SaúdePortugal
| | - Michael Cross
- Department of Medicine 1, Haematology, Cellular Therapy, Hemostaseology and Infectious DiseasesUniversity Medical Center LeipzigLeipzigGermany
| | - Susann Winter
- Medical Clinic I, University Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | | | - Bhavisha A. Patel
- Hematology Branch, National Heart, Lung and Blood InstituteBethesdaMarylandUSA
| | - Emma M. Groarke
- Hematology Branch, National Heart, Lung and Blood InstituteBethesdaMarylandUSA
| | - Daniel H. Wiseman
- Division of Cancer SciencesThe University of ManchesterManchesterUK
- The Christie NHS Foundation TrustManchesterUK
| | - Katy Jones
- Immunophenotyping Laboratory (Synnovis Analytics LLP)Southeast Haematological Malignancy Diagnostic Service, King's College HospitalLondonUK
| | - Lauren Jamieson
- Immunophenotyping Laboratory (Synnovis Analytics LLP)Southeast Haematological Malignancy Diagnostic Service, King's College HospitalLondonUK
| | - Charles Manogaran
- Immunophenotyping Laboratory (Synnovis Analytics LLP)Southeast Haematological Malignancy Diagnostic Service, King's College HospitalLondonUK
| | - Naval Daver
- University of TexasMD Anderson Cancer CenterHouston, TexasUSA
| | - Laura Gallur
- Hematology Department, Vall d'hebron University Hospital, Vall d'hebron Institut of Oncology (VHIO)Vall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Wendy Ingram
- Department of HaematologyUniversity Hospital of WalesCardiffUK
| | - P. Brent Ferrell
- Vanderbilt‐Ingram Cancer Center, Vanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Katja Sockel
- Medical Clinic I, University Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | - Nicolas Dulphy
- INSERM UMR_S1160, Institut de Recherche Saint LouisUniversité Paris CitéParisFrance
- Laboratoire d'Immunologie et d‘Histocompatibilité, Assistance Publique des Hôpitaux de Paris (APHP), Hôpital Saint‐LouisParisFrance
- Institut Carnot OPALE, Institut de Recherche Saint‐Louis, Hôpital Saint‐LouisParisFrance
| | - Nicolas Chapuis
- Université Paris Cité, CNRS, INSERM, Institut CochinParisFrance
- Assistance Publique‐Hôpitaux de Paris Centre, Hôpital CochinParisFrance
| | - Anne S. Kubasch
- Department of Medicine 1, Haematology, Cellular Therapy, Hemostaseology and Infectious DiseasesUniversity Medical Center LeipzigLeipzigGermany
| | - Astrid M. Olsnes
- Section for Hematology, Department of MedicineHaukeland University HospitalBergenNorway
- Department of Clinical ScienceFaculty of Medicine, University of BergenBergenNorway
| | | | | | | | | | - Dominique Bonnet
- Hematopoietic Stem Cell LaboratoryFrancis Crick InstituteLondonUK
| | - Theresia M. Westers
- Department of Hematology, Cancer Center AmsterdamAmsterdam University Medical Centers, location VU University Medical CenterAmsterdamThe Netherlands
| | - Sylvie Freeman
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | - Uta Oelschlaegel
- Medical Clinic I, University Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | - David Valcarcel
- Hematology Department, Vall d'hebron University Hospital, Vall d'hebron Institut of Oncology (VHIO)Vall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Marco G. Raddi
- Myelodysplastic Syndrome Unit, Hematology DivisionAzienda Ospedaliero‐Universitaria Careggi, University of FlorenceFlorenceItaly
| | - Kirsten Grønbæk
- Department of Hematology, RigshospitaletCopenhagen University HospitalCopenhagenDenmark
- Biotech Research and Innovation Center (BRIC)University of CopenhagenCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Michaela Fontenay
- Université Paris Cité, CNRS, INSERM, Institut CochinParisFrance
- Assistance Publique‐Hôpitaux de Paris Centre, Hôpital CochinParisFrance
| | - Sanam Loghavi
- University of TexasMD Anderson Cancer CenterHouston, TexasUSA
| | - Valeria Santini
- Myelodysplastic Syndrome Unit, Hematology DivisionAzienda Ospedaliero‐Universitaria Careggi, University of FlorenceFlorenceItaly
| | - Antonio M. Almeida
- Hematology DepartmentHospital da Luz LisboaLisboaPortugal
- DeaneryFaculdade de Medicina, UCPLisboaPortugal
| | - Jonathan M. Irish
- Cell & Developmental BiologyVanderbilt University School of MedicineNashvilleTennesseeUSA
- Pathology, Microbiology and Immunology, Vanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt‐Ingram Cancer Center, Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | - Neal S. Young
- Hematology Branch, National Heart, Lung and Blood InstituteBethesdaMarylandUSA
| | - Arjan A. van de Loosdrecht
- Department of Hematology, Cancer Center AmsterdamAmsterdam University Medical Centers, location VU University Medical CenterAmsterdamThe Netherlands
| | - Lionel Adès
- Hématologie seniorsHôpital Saint‐Louis, Assistance Publique des Hôpitaux de Paris (APHP)ParisFrance
- Université Paris Cité, CNRS, INSERM, Institut CochinParisFrance
| | - Matteo G. Della Porta
- Humanitas Clinical and Research Center–IRCCS & Department of Biomedical SciencesHumanitas UniversityMilanItaly
| | | | - Uwe Platzbecker
- Department of Medicine 1, Haematology, Cellular Therapy, Hemostaseology and Infectious DiseasesUniversity Medical Center LeipzigLeipzigGermany
| | - Shahram Kordasti
- Comprehensive Cancer Centre, King's CollegeLondonUK
- Department of Clinical and Molecular SciencesUniversità Politecnica delle MarcheAnconaItaly
- Haematology DepartmentGuy's and St Thomas NHS TrustLondonUK
| | | |
Collapse
|
10
|
Zhang L, Deeb G, Deeb KK, Vale C, Peker Barclift D, Papadantonakis N. Measurable (Minimal) Residual Disease in Myelodysplastic Neoplasms (MDS): Current State and Perspectives. Cancers (Basel) 2024; 16:1503. [PMID: 38672585 PMCID: PMC11048433 DOI: 10.3390/cancers16081503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Myelodysplastic Neoplasms (MDS) have been traditionally studied through the assessment of blood counts, cytogenetics, and morphology. In recent years, the introduction of molecular assays has improved our ability to diagnose MDS. The role of Measurable (minimal) Residual Disease (MRD) in MDS is evolving, and molecular and flow cytometry techniques have been used in several studies. In this review, we will highlight the evolving concept of MRD in MDS, outline the various techniques utilized, and provide an overview of the studies reporting MRD and the correlation with outcomes.
Collapse
Affiliation(s)
- Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - George Deeb
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kristin K. Deeb
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Colin Vale
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Deniz Peker Barclift
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nikolaos Papadantonakis
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
11
|
Dimitriou M, Mortera-Blanco T, Tobiasson M, Mazzi S, Lehander M, Högstrand K, Karimi M, Walldin G, Jansson M, Vonlanthen S, Ljungman P, Langemeijer S, Yoshizato T, Hellström-Lindberg E, Woll PS, Jacobsen SEW. Identification and surveillance of rare relapse-initiating stem cells during complete remission after transplantation. Blood 2024; 143:953-966. [PMID: 38096358 PMCID: PMC10950475 DOI: 10.1182/blood.2023022851] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 02/29/2024] Open
Abstract
ABSTRACT Relapse after complete remission (CR) remains the main cause of mortality after allogeneic stem cell transplantation for hematological malignancies and, therefore, improved biomarkers for early prediction of relapse remains a critical goal toward development and assessment of preemptive relapse treatment. Because the significance of cancer stem cells as a source of relapses remains unclear, we investigated whether mutational screening for persistence of rare cancer stem cells would enhance measurable residual disease (MRD) and early relapse prediction after transplantation. In a retrospective study of patients who relapsed and patients who achieved continuous-CR with myelodysplastic syndromes and related myeloid malignancies, combined flow cytometric cell sorting and mutational screening for persistence of rare relapse-initiating stem cells was performed in the bone marrow at multiple CR time points after transplantation. In 25 CR samples from 15 patients that later relapsed, only 9 samples were MRD-positive in mononuclear cells (MNCs) whereas flowcytometric-sorted hematopoietic stem and progenitor cells (HSPCs) were MRD-positive in all samples, and always with a higher variant allele frequency than in MNCs (mean, 97-fold). MRD-positivity in HSPCs preceded MNCs in multiple sequential samples, in some cases preceding relapse by >2 years. In contrast, in 13 patients in long-term continuous-CR, HSPCs remained MRD-negative. Enhanced MRD sensitivity was also observed in total CD34+ cells, but HSPCs were always more clonally involved (mean, 8-fold). In conclusion, identification of relapse-initiating cancer stem cells and mutational MRD screening for their persistence consistently enhances MRD sensitivity and earlier prediction of relapse after allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Marios Dimitriou
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Teresa Mortera-Blanco
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Magnus Tobiasson
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Stefania Mazzi
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Madeleine Lehander
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Kari Högstrand
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mohsen Karimi
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA
| | - Gunilla Walldin
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Monika Jansson
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Sofie Vonlanthen
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Per Ljungman
- Division of Hematology, Department of Medicine, Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Saskia Langemeijer
- Department of Hematology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Tetsuichi Yoshizato
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Eva Hellström-Lindberg
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Petter S. Woll
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Sten Eirik W. Jacobsen
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Haematopoietic Stem Cell Biology Laboratory and MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
12
|
Bashiru M, Macchi S, Forson M, Khan A, Ishtiaq A, Oyebade A, Jalihal A, Ali N, Griffin RJ, Oyelere AK, Hooshmand N, Siraj N. Doxorubicin-Based Ionic Nanomedicines for Combined Chemo-Phototherapy of Cancer. ACS APPLIED NANO MATERIALS 2024; 7:2176-2189. [PMID: 38410412 PMCID: PMC10896075 DOI: 10.1021/acsanm.3c05464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Synergistic combination therapy approach offers lots of options for delivery of materials with anticancer properties, which is a very promising strategy to treat a variety of malignant lesions with enhanced therapeutic efficacy. The current study involves a detailed investigation of combination ionic nanomedicines where a chemotherapeutic drug is coupled with a photothermal agent to attain dual mechanisms (chemotherapy (chemo) and photothermal therapy (PTT)) to improve the drug's efficacy. An FDA-approved Doxorubicin hydrochloride (DOX·HCl) is electrostatically attached with a near-infrared cyanine dye (ICG, IR783, and IR820), which serves as a PTT drug using ionic liquid chemistry to develop three ionic material (IM)-based chemo-PTT drugs. Carrier-free ionic nanomedicines (INMs) are derived from ionic materials (IMs). The photophysical properties of the developed combination IMs and their INMs were studied in depth. The phototherapeutic efficiency of the combination drugs was evaluated by measuring the photothermal conversion efficiency and singlet-oxygen quantum yield. The improved photophysical properties of the combination nanomedicines in comparison to their parent compounds significantly enhanced INMs' photothermal efficiency. Cellular uptake, dark and light toxicity studies, and cell death mechanisms of the chemo-PTT nanoparticles were also studied in vitro. The combination INMs exhibited enhanced cytotoxicity compared to their respective parent compounds. Moreover, the apoptosis cell death mechanism was almost doubled for combination nanomedicine than the free DOX, which is attributed to enhanced cellular uptake. Examination of the combination index and improved in vitro cytotoxicity results revealed a great synergy between chemo and PTT drugs in the developed combination nanomedicines.
Collapse
Affiliation(s)
- Mujeebat Bashiru
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Samantha Macchi
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Mavis Forson
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Amna Khan
- Department of Chemistry, University of Arkansas at Fayetteville, Fayetteville, Arkansas 72701, United States
| | - Arisha Ishtiaq
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Adeniyi Oyebade
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Amanda Jalihal
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Nawab Ali
- Department of Biology, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Robert J Griffin
- Department of Radiation Oncology, Arkansas Nanomedicine Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Nasrin Hooshmand
- Laser Dynamics Laboratory, School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Noureen Siraj
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| |
Collapse
|
13
|
Abstract
Myelodysplastic syndromes/neoplasms (MDS) are a heterogeneous class of hematopoietic stem cell neoplasms characterized by ineffective hematopoiesis leading to peripheral cytopenias. This group of diseases is typically diagnosed using a combination of clinical, morphologic, and genetic criteria. Many studies have described the value of multiparametric flow cytometry (MFC) in the diagnosis, classification, and prognostication of MDS. This review summarizes the approach to MDS diagnosis and immunophenotypic characterization using MFC and describes the current state while highlighting future opportunities and potential pitfalls.
Collapse
Affiliation(s)
- Xueyan Chen
- Translational Science and Therapeutics Division, Fred Hutch Cancer Center, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, 825 Eastlake Avenue East, Seattle, WA 98109, USA
| | - Ulrika Johansson
- SI-HMDS, Haematology, UHBW NHS Foundation Trust, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Sindhu Cherian
- Department of Laboratory Medicine and Pathology, University of Washington, 825 Eastlake Avenue East, Seattle, WA 98109, USA.
| |
Collapse
|
14
|
Saft L. The role of flow cytometry in the classification of myeloid disorders. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:164-175. [PMID: 37991530 PMCID: PMC10739577 DOI: 10.1007/s00292-023-01272-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 11/23/2023]
Abstract
The World Health Organization classification (WHO-HAEM5) and the International Consensus Classification (ICC 2022) of myeloid neoplasms are based on the integration of clinical, morphologic, immunophenotypic, and genomic data. Flow cytometric immunophenotyping (FCIP) allows the identification, enumeration, and characterization of hematopoietic cells, and is therefore a powerful tool in the diagnosis, classification, and monitoring of hematological neoplasms. The vast majority of flow cytometry (FCM) studies in chronic myeloid neoplasms focus on its role in myelodysplastic neoplasms (MDS). FCM can also be helpful for the assessment of myeloproliferative neoplasms (MPN) and MDS/MPN, including the early detection of evolving myeloid or lymphoid blast crisis and the characterization of monocytic subsets. The classification of acute myeloid leukemia (AML) is primarily based on cytogenetic and molecular findings; however, FCIP is needed for subclassification of AML, not otherwise specified (NOS; ICC)/AML defined by differentiation (WHO-HAEM5). The main role of FCM in AML remains in making a rapid diagnosis and as a tool for measurable residual disease monitoring. Machine learning and artificial intelligence approaches can be used to analyze and classify FCM data. This article, based on an invited lecture at the 106th Annual Meeting of the German Society of Pathology in 2023, reviews the role of FCM in the classification of myeloid neoplasms, including recent publications on the application of artificial intelligence.
Collapse
Affiliation(s)
- Leonie Saft
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital and Institute, 171 76, Stockholm, Sweden.
| |
Collapse
|
15
|
Shameli A, Dharmani-Khan P, Auer I, Shabani-Rad MT. Deep immunophenotypic analysis of the bone marrow progenitor cells in myelodysplastic syndromes. Leuk Res 2023; 134:107401. [PMID: 37774446 DOI: 10.1016/j.leukres.2023.107401] [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: 02/20/2023] [Revised: 08/23/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND Diagnosis of myelodysplastic syndromes (MDS) is often challenging and requires integration of clinical, morphologic, cytogenetics and molecular information. Flow cytometry immunophenotyping (FCIP) can support the diagnosis by demonstration of numerical and immunophenotypic abnormalities of progenitor and maturing myelomonocytic and erythroid populations. We have previously shown that comprehensive immunophenotypic analysis of the progenitor population is valuable in the diagnosis of MDS and myelodysplastic/myeloproliferative neoplasms (MDS/MPN). This study was designed to improve the analysis method and confirm its value in a larger cohort of patients. METHODS FCIP of bone marrow samples from 105 patients with cytopenia(s) (with or without leukocytosis) and clinical concern for MDS or MDS/MPN was performed using a single-tube/10-color/13-marker assay. A modified analysis approach was used to obtain 11 progenitor parameters and 2 myelomonocytic parameters. RESULTS Significantly higher number of abnormalities were identified in MDS and MDS/MPN cases when compared to cytopenic patients not meeting the diagnostic criteria for MDS (Non-MDS). A FCIP score that combined the 13 parameters showed a sensitivity of 89.8% and specificity of 93.5% for the diagnosis of MDS and MDS/MPN. The sensitivity was 100% for both MDS/MPN and higher-risk MDS, and 81.3% for lower-risk MDS. CONCLUSION This study confirms that detailed immunophenotypic analysis of the progenitor population is powerful in the diagnosis of MDS and MDS/MPN. The combination of markers used in the panel allowed for evaluation of two relatively new parameters, namely myeloid progenitor heterogeneity and stem cell aberrancy, which improved the sensitivity of the assay for lower-risk MDS.
Collapse
Affiliation(s)
- Afshin Shameli
- Department of Laboratory Medicine and Pathology, University of Washington, WA, United States.
| | - Poonam Dharmani-Khan
- Division of Hematopathology, Alberta Precision Laboratories, South Zone, and Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Iwona Auer
- Division of Hematopathology, Alberta Precision Laboratories, South Zone, and Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Meer-Taher Shabani-Rad
- Division of Hematopathology, Alberta Precision Laboratories, South Zone, and Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
16
|
Guarnera L, Fabiani E, Attrotto C, Hajrullaj H, Cristiano A, Latagliata R, Fenu S, Buccisano F, Irno-Consalvo M, Conti C, Voso MT, Maurillo L. Reliability of flow-cytometry in diagnosis and prognostic stratification of myelodysplastic syndromes: correlations with morphology and mutational profile. Ann Hematol 2023; 102:3015-3023. [PMID: 37535147 PMCID: PMC10567902 DOI: 10.1007/s00277-023-05384-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
Diagnosis and prognostic stratification of myelodysplastic syndromes (MDS) have been complemented by new techniques, including flow cytometry and NGS. To analyze the relationship between molecular and cytofluorimetric data, we enrolled in this retrospective study, 145 patients, including 106 diagnosed with MDS and 39 controls. At disease onset, immunophenotypic (IF), cytogenetic tests, and cytomorphological (CM) examination on bone marrow were carried out in all patients, while NGS was performed in 58 cases. Ogata score presented a specificity of 100% and a sensitivity of 59%. The detection of at least two phenotypic aberrancies in Ogata negative patients increased the sensitivity to 83% and specificity to 87%. Correlations were identified between IF aberrancies and mutations, including positive Ogata>2 and mutations in SRSF2 (p=0.035), CD15 and U2AF1 (0.032), CD56 and DNMT3A (p=0.042), and CD38 and TP53 (p=0.026). In multivariate analysis, U2AF1 mutations, associated with del(20q) and/or abnormalities of chromosome 7 (group 4 as defined by the EuroMDS score), significantly correlated with an inferior overall survival (p=0.019). These parameters and Ogata score>2 also showed a significant correlation with inferior event-free survival (p=0.023 and p=0.041, respectively). Both CM and FC features correlated with prognosis and mutational patterns. In an integrated MDS work-up, these tools may guide indications for mutational screening for optimal risk stratification.
Collapse
Affiliation(s)
- Luca Guarnera
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Emiliano Fabiani
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - Cristina Attrotto
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Hajro Hajrullaj
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Antonio Cristiano
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | | | - Susanna Fenu
- Haematology Department, San Giovanni-Addolorata Hospital, Rome, Italy
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Maria Irno-Consalvo
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Consuelo Conti
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy.
- Neuro-Oncohematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy.
| | - Luca Maurillo
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
17
|
Jurado R, Huguet M, Xicoy B, Cabezon M, Jimenez-Ponce A, Quintela D, De La Fuente C, Raya M, Vinets E, Junca J, Julià-Torras J, Zamora L, Oriol A, Navarro JT, Calvo X, Sorigue M. Optimization of monocyte gating to quantify monocyte subsets for the diagnosis of chronic myelomonocytic leukemia. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:319-330. [PMID: 36448679 DOI: 10.1002/cyto.b.22106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/03/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND The presence of >94% classical monocytes (MO1, CD14++/CD16-) in peripheral blood (PB) has an excellent performance for the diagnosis of chronic myelomonocytic leukemia (CMML). However, the monocyte gating strategy is not well defined. The objective of the study was to compare monocyte gating strategies and propose an optimal one. METHODS This is a prospective, single center study assessing monocyte subsets in PB. First, we compared monocyte subsets using 13 monocyte gating strategies in 10 samples. Then we developed our own 10 color tube and tested it on 124 patients (normal white blood cell counts, reactive monocytosis, CMML and a spectrum of other myeloid malignancies). Both conventional and computational (FlowSOM) analyses were used. RESULTS Comparing different monocyte gating strategies, small but significant differences in %MO1 and percentually large differences in %MO3 (nonclassical monocytes) were found, suggesting that the monocyte gating strategy can impact monocyte subset quantification. Then, we designed a 10-color tube for this purpose (CD45/CD33/CD14/CD16/CD64/CD86/CD300/CD2/CD66c/CD56) and applied it to 124 patients. This tube allowed proper monocyte gating even in highly abnormal PB. Computational analysis found a higher %MO1 and lower %MO3 compared to conventional analysis. However, differences between conventional and computational analysis in both MO1 and MO3 were globally consistent and only minimal differences were observed when comparing the ranking of patients according to %MO1 or %MO3 obtained with the conventional versus the computational approach. CONCLUSIONS The choice of monocyte gating strategy appears relevant for the monocyte subset distribution test. Our 10-color proposal allowed satisfactory monocyte gating even in highly abnormal PB. Computational analysis seems promising to increase reproducibility in monocyte subset quantification.
Collapse
Affiliation(s)
- Rebeca Jurado
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Maria Huguet
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Blanca Xicoy
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Marta Cabezon
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Ari Jimenez-Ponce
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - David Quintela
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Cristina De La Fuente
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Minerva Raya
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Esther Vinets
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Jordi Junca
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | | | - Lurdes Zamora
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Albert Oriol
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Jose-Tomas Navarro
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| | - Xavier Calvo
- Laboratori de Citologia Hematològica, Servei de Patologia, Grup de Recerca Translacional en Neoplàsies Hematològiques (GRETNHE), IMIM Hospital del Mar Research Institute, Barcelona, Spain
| | - Marc Sorigue
- Department of Hematology, ICO-IJC-Hospital Germans Trias i Pujol, LUMN, UAB, Badalona, Spain
| |
Collapse
|
18
|
Sorigue M. Diagnosis of erythroid dysplasia by flow cytometry: a review. Expert Rev Hematol 2023; 16:1049-1062. [PMID: 38018383 DOI: 10.1080/17474086.2023.2289534] [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: 08/18/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION The diagnosis of myelodysplastic syndrome (MDS) is complex. Flow cytometric analysis of the myelomonocytic compartment can be helpful, but it is highly subjective and reproducibility by non-specialized groups is unclear. Analysis of the erythroid lineage by flow cytometry is emerging as potentially more reproducible and easier to conduct, while keeping a high diagnostic performance. AREAS COVERED We review the evidence in this area, including 1) the use of well-established markers - CD71 and CD36 - and other less well-established markers and parameters; 2) the use of flow cytometric scores for the erythroid lineage; and 3) additional aspects, including the emergence of computational tools and the roles of flow cytometry beyond diagnosis. Finally, we discuss the limitations with the current evidence, including 1) the impact of the sample processing protocol and reagents on the results, 2) the lack of a standard gating strategy, and 3) conceptualization and design issues in the available publications. EXPERT OPINION We end by offering our recommendations for the current use - and our personal take on the value - of the analysis of erythroid lineage by flow cytometry.
Collapse
Affiliation(s)
- Marc Sorigue
- Medical Department, Trialing Health, Barcelona, Spain
| |
Collapse
|
19
|
Bachas C, Duetz C, van Spronsen MF, Verhoeff J, Garcia Vallejo JJ, Jansen JH, Cloos J, Westers TM, van de Loosdrecht AA. Characterization of myelodysplastic syndromes hematopoietic stem and progenitor cells using mass cytometry. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:128-140. [PMID: 35289472 DOI: 10.1002/cyto.b.22066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/13/2022] [Accepted: 02/28/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Myelodysplastic syndromes (MDS) at risk of transformation to acute myeloid leukemia (AML) are difficult to identify. The bone marrows of MDS patients harbor specific hematopoietic stem and progenitor cell (HSPC) abnormalities that may be associated with sub-types and risk-groups. Leukemia-associated characteristics of such cells may identify MDS patients at risk of progression to AML and provide insight in the pathobiology of MDS. METHODS Bone marrow samples from healthy donors (n = 10), low risk (n = 12) and high risk (n = 13) MDS patients were collected, in addition, AML samples for 5 out of 6 MDS patients that progressed. Mass cytometry was applied to assess expression of stem cell subset and leukemia-associated immunophenotype markers. RESULTS We analyzed the data using FlowSOM to cluster cells with similar expression of 10 commonly used stem cell markers. Metaclusters (n = 20) of these clusters represented populations of cells with a related phenotype, largely resembling known stem cell subsets. Within specific subsets, intra-cellular expression levels of pCREB, IkBα, or pS6 differed significantly between healthy bone marrow (HBM) and MDS or consecutive secondary AML samples. CD34, CD44, and CD49f expression was significantly increased in high risk MDS and AML-associated metaclusters. We identified MDS/sAML cells with aberrant phenotypes when compared to HBM. Such cells were observed in clusters of both primary MDS and secondary AML samples. CONCLUSIONS High-dimensional mass cytometry and computational data analyses enabled characterization of HSPC subsets in MDS and identification of leukemia stem cell populations based on their immunophenotype. Stem cells in MDS that display leukemia-associated features may predict the risk of developing AML.
Collapse
Affiliation(s)
- Costa Bachas
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Carolien Duetz
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Margot F van Spronsen
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jan Verhoeff
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Juan J Garcia Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Joop H Jansen
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Theresia M Westers
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Arjan A van de Loosdrecht
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Johansson U, McIver-Brown N, Cullen M, Duetz C, Dunlop A, Oelschlägel U, Psarra K, Subirá D, Westers TM. The flow cytometry myeloid progenitor count: A reproducible parameter for diagnosis and prognosis of myelodysplastic syndromes. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:115-127. [PMID: 34931733 DOI: 10.1002/cyto.b.22048] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 12/04/2021] [Accepted: 12/09/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND The bone marrow blast count is central to the diagnosis and monitoring of myelodysplastic syndromes (MDS). It is an independent risk factor for worse prognosis whether based on the morphology blast count or the flow cytometry (FC) myeloid progenitor (MyP) count. It is a principal population in FC MDS analysis also because once defined; it provides significant contributions to the overall FC MDS score. METHODS We elected to investigate inter-analyst agreement for the most fundamental parameter of the FC MDS diagnostic score: the MyP count. A common gating strategy was agreed and used by seven cytometrists for blind analysis of 34 routine bone marrows sent for MDS work-up. Additionally, we compared the results with a computational approach. RESULTS Concordance was excellent: Intraclass correlation was 0.993 whether measuring %MyP of total cells or CD45+ cells, and no significant difference was observed between files from different centers or for samples with abnormal MyP phenotypes. Computational and manual results were similar. Applying the common strategy to individual laboratories' control cohorts produced similar MyP reference ranges across centers. CONCLUSION The FC MyP count offers a reliable diagnostic and prognostic measurement in MDS. The use of manual and computational approaches side by side may allow for optimizing both strategies. Considering its known prognostic power, the MyP count could be considered a useful and reliable addition to existing prognostic scoring systems.
Collapse
Affiliation(s)
- Ulrika Johansson
- Haematological Malignancy Diagnostic Service, University Hospitals and Weston NHS Foundation Trust, Bristol, UK
| | - Neil McIver-Brown
- Molecular Pathology Department, Royal Bournemouth Hospital, University Hospitals Dorset NHS Foundation Trust, Bournemouth, UK
| | - Matthew Cullen
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Carolien Duetz
- Department of Haematology, Amsterdam University Medical Centres, VU University Medical Center, Amsterdam, The Netherlands
| | - Alan Dunlop
- Haematological Malignancy Diagnostic Centre, King's College Hospital NHS Foundation Trust, London, UK
| | - Uta Oelschlägel
- Department of Haematology, Medical Clinic and Policlinic I, University Hospital of TU Dresden, Dresden, Germany
| | - Katherina Psarra
- Department of Immunology and Histocompatibility, Evangelismos Hospital, Athens, Greece
| | - Dolores Subirá
- Department of Haematology, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | - Theresia M Westers
- Department of Hematology, Amsterdam University Medical Centers, Location VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Porwit A, Béné MC, Duetz C, Matarraz S, Oelschlaegel U, Westers TM, Wagner-Ballon O, Kordasti S, Valent P, Preijers F, Alhan C, Bellos F, Bettelheim P, Burbury K, Chapuis N, Cremers E, Della Porta MG, Dunlop A, Eidenschink-Brodersen L, Font P, Fontenay M, Hobo W, Ireland R, Johansson U, Loken MR, Ogata K, Orfao A, Psarra K, Saft L, Subira D, Te Marvelde J, Wells DA, van der Velden VHJ, Kern W, van de Loosdrecht AA. Multiparameter flow cytometry in the evaluation of myelodysplasia: Analytical issues: Recommendations from the European LeukemiaNet/International Myelodysplastic Syndrome Flow Cytometry Working Group. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:27-50. [PMID: 36537621 PMCID: PMC10107708 DOI: 10.1002/cyto.b.22108] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/20/2022] [Accepted: 11/29/2022] [Indexed: 01/18/2023]
Abstract
Multiparameter flow cytometry (MFC) is one of the essential ancillary methods in bone marrow (BM) investigation of patients with cytopenia and suspected myelodysplastic syndrome (MDS). MFC can also be applied in the follow-up of MDS patients undergoing treatment. This document summarizes recommendations from the International/European Leukemia Net Working Group for Flow Cytometry in Myelodysplastic Syndromes (ELN iMDS Flow) on the analytical issues in MFC for the diagnostic work-up of MDS. Recommendations for the analysis of several BM cell subsets such as myeloid precursors, maturing granulocytic and monocytic components and erythropoiesis are given. A core set of 17 markers identified as independently related to a cytomorphologic diagnosis of myelodysplasia is suggested as mandatory for MFC evaluation of BM in a patient with cytopenia. A myeloid precursor cell (CD34+ CD19- ) count >3% should be considered immunophenotypically indicative of myelodysplasia. However, MFC results should always be evaluated as part of an integrated hematopathology work-up. Looking forward, several machine-learning-based analytical tools of interest should be applied in parallel to conventional analytical methods to investigate their usefulness in integrated diagnostics, risk stratification, and potentially even in the evaluation of response to therapy, based on MFC data. In addition, compiling large uniform datasets is desirable, as most of the machine-learning-based methods tend to perform better with larger numbers of investigated samples, especially in such a heterogeneous disease as MDS.
Collapse
Affiliation(s)
- Anna Porwit
- Division of Oncology and Pathology, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Marie C Béné
- Hematology Biology, Nantes University Hospital, CRCINA Inserm 1232, Nantes, France
| | - Carolien Duetz
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sergio Matarraz
- Cancer Research Center (IBMCC-USAL/CSIC), Department of Medicine and Cytometry Service, Institute for Biomedical Research of Salamanca (IBSAL) and CIBERONC, University of Salamanca, Salamanca, Spain
| | - Uta Oelschlaegel
- Department of Internal Medicine, University Hospital Carl-Gustav-Carus, TU Dresden, Dresden, Germany
| | - Theresia M Westers
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Orianne Wagner-Ballon
- Department of Hematology and Immunology, Assistance Publique-Hôpitaux de Paris, University Hospital Henri Mondor, Créteil, France
- Inserm U955, Université Paris-Est Créteil, Créteil, France
| | | | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Frank Preijers
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Canan Alhan
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Peter Bettelheim
- Department of Hematology, Ordensklinikum Linz, Elisabethinen, Linz, Austria
| | - Kate Burbury
- Department of Haematology, Peter MacCallum Cancer Centre, & University of Melbourne, Melbourne, Australia
| | - Nicolas Chapuis
- Laboratory of Hematology, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Cochin Hospital, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR, Université de Paris, Paris, France
| | - Eline Cremers
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Matteo G Della Porta
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Alan Dunlop
- Department of Haemato-Oncology, Royal Marsden Hospital, London, UK
| | | | - Patricia Font
- Department of Hematology, Hospital General Universitario Gregorio Marañon-IiSGM, Madrid, Spain
| | - Michaela Fontenay
- Laboratory of Hematology, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Cochin Hospital, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR, Université de Paris, Paris, France
| | - Willemijn Hobo
- Department of Internal Medicine I, Division of Hematology & Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Robin Ireland
- Department of Haematology and SE-HMDS, King's College Hospital NHS Foundation Trust, London, UK
| | - Ulrika Johansson
- Laboratory Medicine, SI-HMDS, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | | | - Kiyoyuki Ogata
- Metropolitan Research and Treatment Centre for Blood Disorders (MRTC Japan), Tokyo, Japan
| | - Alberto Orfao
- Cancer Research Center (IBMCC-USAL/CSIC), Department of Medicine and Cytometry Service, Institute for Biomedical Research of Salamanca (IBSAL) and CIBERONC, University of Salamanca, Salamanca, Spain
| | - Katherina Psarra
- Department of Immunology - Histocompatibility, Evangelismos Hospital, Athens, Greece
| | - Leonie Saft
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital and Institute Solna, Stockholm, Sweden
| | - Dolores Subira
- Department of Hematology, Flow Cytometry Unit, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | - Jeroen Te Marvelde
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Vincent H J van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Arjan A van de Loosdrecht
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Wang W, Khoury JD. Where diagnosis for myelodysplastic neoplasms (MDS) stands today and where it will go: The role of flow cytometry in evaluation of MDS. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:12-14. [PMID: 36573832 DOI: 10.1002/cyto.b.22110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022]
Affiliation(s)
- Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joseph D Khoury
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
23
|
Kern W, Westers TM, Bellos F, Bene MC, Bettelheim P, Brodersen LE, Burbury K, Chu SC, Cullen M, Porta MD, Dunlop AS, Johansson U, Matarraz S, Oelschlaegel U, Ogata K, Porwit A, Preijers F, Psarra K, Saft L, Subirá D, Weiß E, van der Velden VHJ, van de Loosdrecht A. Multicenter prospective evaluation of diagnostic potential of flow cytometric aberrancies in myelodysplastic syndromes by the ELN iMDS flow working group. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:51-65. [PMID: 36416672 DOI: 10.1002/cyto.b.22105] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 10/31/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Myelodysplastic syndromes (MDS) represent a diagnostic challenge. This prospective multicenter study was conducted to evaluate pre-defined flow cytometric markers in the diagnostic work-up of MDS and chronic myelomonocytic leukemia (CMML). METHODS Thousand six hundred and eighty-two patients with suspected MDS/CMML were analyzed by both cytomorphology according to WHO 2016 criteria and flow cytometry according to ELN recommendations. Flow cytometric readout was categorized 'non-MDS' (i.e. no signs of MDS/CMML and limited signs of MDS/CMML) and 'in agreement with MDS' (i.e., in agreement with MDS/CMML). RESULTS Flow cytometric readout categorized 60% of patients in agreement with MDS, 28% showed limited signs of MDS and 12% had no signs of MDS. In 81% of cases flow cytometric readouts and cytomorphologic diagnosis correlated. For high-risk MDS, the level of concordance was 92%. A total of 17 immunophenotypic aberrancies were found independently related to MDS/CMML in ≥1 of the subgroups of low-risk MDS, high-risk MDS, CMML. A cut-off of ≥3 of these aberrancies resulted in 80% agreement with cytomorphology (20% cases concordantly negative, 60% positive). Moreover, >3% myeloid progenitor cells were significantly associated with MDS (286/293 such cases, 98%). CONCLUSION Data from this prospective multicenter study led to recognition of 17 immunophenotypic markers allowing to identify cases 'in agreement with MDS'. Moreover, data emphasizes the clinical utility of immunophenotyping in MDS diagnostics, given the high concordance between cytomorphology and the flow cytometric readout. Results from the current study challenge the application of the cytomorphologically defined cut-off of 5% blasts for flow cytometry and rather suggest a 3% cut-off for the latter.
Collapse
Affiliation(s)
| | - Theresia M Westers
- Department of Hematology, Amsterdam University Medical Centers, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | | | - Peter Bettelheim
- Department of Hematology, Elisabethinen Hospital, Linz, Upper Austria, Austria
| | | | - Kate Burbury
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Sung-Chao Chu
- Department of Hematology and Oncology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Matthew Cullen
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Matteo Della Porta
- Department of Biomedical Sciences, IRCCS Humanitas Research Hospital, Humanitas University, Milan, Italy
| | | | - Ulrika Johansson
- Laboratory Medicine, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Sergio Matarraz
- Cytometry Service (NUCLEUS), Department of Medicine and IBSAL, Cancer Research Center (IBMCC, University of Salamanca-CSIC), Salamanca, Spain and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Salamanca, Spain
| | - Uta Oelschlaegel
- Department of Internal Medicine, University Hospital of Technical University Dresden, Dresden, Germany
| | - Kiyoyuki Ogata
- Metropolitan Research and Treatment Centre for Blood Disorders (MRTC Japan), Tokyo, Japan
| | - Anna Porwit
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Frank Preijers
- Department of Laboratory Medicine, Laboratory of Hematology, Radboudumc, Nijmegen, The Netherlands
| | - Katherina Psarra
- Immunology Histocompatibility Department, Evangelismos Hospital, Athens, Greece
| | - Leonie Saft
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital and Institute, Stockholm, Sweden
| | - Dolores Subirá
- Department of Medical Immunology, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | | | - Vincent H J van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Arjan van de Loosdrecht
- Department of Hematology, Amsterdam University Medical Centers, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Brunner AM, Leitch HA, van de Loosdrecht AA, Bonadies N. Management of patients with lower-risk myelodysplastic syndromes. Blood Cancer J 2022; 12:166. [PMID: 36517487 PMCID: PMC9751093 DOI: 10.1038/s41408-022-00765-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of hematopoietic stem cell disorders characterized by ineffective hematopoiesis with abnormal blood cell development (dysplasia) leading to cytopenias and an increased risk for progression to acute myeloid leukemia (AML). Patients with MDS can generally be classified as lower- (LR-MDS) or higher-risk (HR-MDS). As treatment goals for patients with LR-MDS and those with HR-MDS differ significantly, appropriate diagnosis, classification, and follow-up are critical for correct disease management. In this review, we focus on the diagnosis, prognosis, and treatment options, as well as the prediction of the disease course and monitoring of treatment response in patients with LR-MDS. We discuss how next-generation sequencing, increasing knowledge on mechanisms of MDS pathogenesis, and novel therapies may change the current treatment landscape in LR-MDS and why structured assessments of responses, toxicities, and patient-reported outcomes should be incorporated into routine clinical practice.
Collapse
Affiliation(s)
| | - Heather A Leitch
- Hematology, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Arjan A van de Loosdrecht
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, VU University Amsterdam, Amsterdam, Netherlands
| | - Nicolas Bonadies
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Sędek Ł, Flores-Montero J, van der Sluijs A, Kulis J, te Marvelde J, Philippé J, Böttcher S, Bitter M, Caetano J, van der Velden VHJ, Sonneveld E, Buracchi C, Santos AH, Lima M, Szczepański T, van Dongen JJM, Orfao A. Impact of Pre-Analytical and Analytical Variables Associated with Sample Preparation on Flow Cytometric Stainings Obtained with EuroFlow Panels. Cancers (Basel) 2022; 14:cancers14030473. [PMID: 35158741 PMCID: PMC8833630 DOI: 10.3390/cancers14030473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Objective interpretation of flow cytometry may be hampered by a lack of standardized sample preparation procedures. The EuroFlow consortium conducted a series of experiments to determine the potential impact of different pre-analytical and analytical factors on the variability of results in terms of relative cell populations distribution and marker expression levels. The experiments were performed on healthy donors and patients with different hematological malignancies (e.g., acute leukemia, lymphoma, multiple myeloma, and myelodysplastic syndrome) to mimic real-world clinical settings. Overall, the results showed that sample storage conditions, anticoagulant use, and sample processing protocol might need to be tailored for sample and cell type(s), as well as to the specific markers evaluated. However, defining of well-balanced boundaries for storage time to 24 h, staining-acquisition delay to 3 h, and choosing a washing buffer of pH within the range of 7.2 to 7.8 would be a valid recommendation for most applications and circumstances described herein. Abstract Objective interpretation of FC results may still be hampered by limited technical standardization. The EuroFlow consortium conducted a series of experiments to determine the impact of different variables on the relative distribution and the median fluorescence intensity (MFI) of markers stained on different cell populations, from both healthy donors and patients’ samples with distinct hematological malignancies. The use of different anticoagulants; the time interval between sample collection, preparation, and acquisition; pH of washing buffers; and the use of cell surface membrane-only (SM) vs. cell surface plus intracytoplasmic (SM+CY) staining protocols, were evaluated. Our results showed that only monocytes were represented at higher percentages in EDTA- vs. heparin-anticoagulated samples. Application of SM or SM+CY protocols resulted in slight differences in the percentage of neutrophils and debris determined only with particular antibody combinations. In turn, storage of samples for 24 h at RT was associated with greater percentage of debris and cell doublets when the plasma cell disorder panel was used. Furthermore, 24 h storage of stained cells at RT was selectively detrimental for MFI levels of CD19 and CD45 on mature B- and T-cells (but not on leukemic blasts, clonal B- and plasma cells, neutrophils, and NK cells). The obtained results showed that the variables evaluated might need to be tailored for sample and cell type(s) as well as to the specific markers compared; however, defining of well-balanced boundaries for storage time, staining-to-acquisition delay, and pH of washing buffer would be a valid recommendation for most applications and circumstances described herein.
Collapse
Affiliation(s)
- Łukasz Sędek
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice (SUM), 41-808 Zabrze, Poland;
| | - Juan Flores-Montero
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca (USAL), 37007 Salamanca, Spain; (J.F.-M.); (J.J.M.v.D.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Center of Biomedical Network Research in Cancer (CIBER ONC), Carlos III Institute of Health, 28029 Madrid, Spain
| | - Alita van der Sluijs
- Department of Immunology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands;
| | - Jan Kulis
- Department of Pediatric Hematology and Oncology, Medical University of Silesia in Katowice (SUM), 41-800 Zabrze, Poland; (J.K.); (T.S.)
| | - Jeroen te Marvelde
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (J.t.M.); (V.H.J.v.d.V.)
| | - Jan Philippé
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Sebastian Böttcher
- Special Hematology Laboratory, Medical Clinic III, Hematology, Oncology and Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Marieke Bitter
- European Scientific Foundation for Laboratory Hemato Oncology (ESLHO), 2333 ZA Leiden, The Netherlands;
| | - Joana Caetano
- Clinical Flow, Hemato-Oncology Unit, Champalimaud Foundation, 1400-038 Lisboa, Portugal;
| | - Vincent H. J. van der Velden
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (J.t.M.); (V.H.J.v.d.V.)
| | - Edwin Sonneveld
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
| | - Chiara Buracchi
- Pediatric Clinic of Milano-Bicocca, Tettamanti Research Center, Monza (TRC), 20900 Monza, Italy;
| | - Ana Helena Santos
- Department of Hematology, Central Hospital of Porto (CHP), 4099-001 Porto, Portugal; (A.H.S.); (M.L.)
| | - Margarida Lima
- Department of Hematology, Central Hospital of Porto (CHP), 4099-001 Porto, Portugal; (A.H.S.); (M.L.)
| | - Tomasz Szczepański
- Department of Pediatric Hematology and Oncology, Medical University of Silesia in Katowice (SUM), 41-800 Zabrze, Poland; (J.K.); (T.S.)
| | - Jacques J. M. van Dongen
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca (USAL), 37007 Salamanca, Spain; (J.F.-M.); (J.J.M.v.D.)
- Department of Immunology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands;
| | - Alberto Orfao
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca (USAL), 37007 Salamanca, Spain; (J.F.-M.); (J.J.M.v.D.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Center of Biomedical Network Research in Cancer (CIBER ONC), Carlos III Institute of Health, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-923-294-811
| |
Collapse
|