1
|
Pedreira CE, Lecrevisse Q, Fluxa R, Verde J, Barrena S, Flores-Montero J, Fernandez P, Morf D, van der Velden VHJ, Mejstrikova E, Caetano J, Burgos L, Böttcher S, van Dongen JJM, Orfao A. Comparison between five pattern-based approaches for automated diagnostic classification of mature/peripheral B-cell neoplasms based on standardized EuroFlow flow cytometry immunophenotypic data. Comput Biol Med 2025; 192:110194. [PMID: 40300296 DOI: 10.1016/j.compbiomed.2025.110194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/11/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
Flow cytometry immunophenotyping is critical for the diagnostic classification of mature/peripheral B-cell neoplasms/B-cell chronic lymphoproliferative disorders (B-CLPD). Quantitative driven classification approaches applied to multiparameter flow cytometry immunophenotypic data can be used to extract maximum information from a multidimensional space created by individual parameters (e.g., immunophenotypic markers), for highly accurate and automated classification of individual patient (sample) data. Here, we developed and compared five diagnostic classification algorithms, based on a large set of EuroFlow multicentric flow cytometry data files from a cohort 659 B-CLPD patients. These included automatic population separators based on Principal Component Analysis (PCA), Canonical Variate Analysis (CVA), Neighbourhood Component Analysis (NCA), Support Vector Machine algorithms (SVM) and a variant of the CA(Canonical Analysis) algorithm, in which the number of SDs (Standard Deviations) varied for each of the comparisons of different pairs of diseases (CA-vSD). All five classification approaches are based on direct prospective interrogation of individual B-CLPD patients against the EuroFlow flow cytometry B-CLPD database composed of tumor B-cells of 659 individual patients stained in an identical way and classified a priori by the World Health Organization (WHO) criteria into nine diagnostic categories. Each classification approach was evaluated in parallel in terms of accuracy (% properly classified cases), precision (multiple or single diagnosis/case) and coverage (% cases with a proposed diagnosis). Overall, average rates of correct diagnosis (for the nine B-CLPD diagnostic entities) of between 58.9 % and 90.6 % were obtained with the five algorithms, with variable percentages of cases being either misclassified (4.1 %-14.0 %) or unclassifiable (0.3 %-37.0 %). Automatic population separators based on CA, SVM and PCA showed a high average level of correctness (90.6 %, 86.8 %, and 86.0 %, respectively). Nevertheless, this was at the expense of proposing a considerable number of multiple diagnoses for a significant proportion of the test cases (54.5 %, 53.5 %, and 49.6 %, respectively). The CA-vSD algorithm generated the smaller average misclassification rate (4.1 %), but with 37.0 % of cases for which no diagnosis was proposed. In contrast, the NCA algorithm left only 2.7 % of cases without an associated diagnosis but misclassified 14.0 %. Among correctly classified cases (83.3 % of total), 91.2 % had a single proposed diagnosis, 8.6 % had two possible diagnoses, and 0.2 % had three. We demonstrate that the proposed AI algorithms provide an acceptable level of accuracy for the diagnostic classification of B-CLPD patients and, in general, surpass other algorithms reported in the literature.
Collapse
Affiliation(s)
- C E Pedreira
- Systems and Computing Department (PESC), COPPE, Federal University of Rio de Janeiro (UFRJ), Brazil.
| | - Q Lecrevisse
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - R Fluxa
- Cytognos SL, Salamanca, Spain
| | - J Verde
- Cytognos SL, Salamanca, Spain
| | - S Barrena
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - J Flores-Montero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - P Fernandez
- Institute for Laboratory Medicine, Kantonsspital Aarau AG, Aarau, Switzerland
| | - D Morf
- Institute for Laboratory Medicine, Kantonsspital Aarau AG, Aarau, Switzerland
| | - V H J van der Velden
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - E Mejstrikova
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Charles University, Prague, Czechia
| | - J Caetano
- Secção de Citometria de Fluxo, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - L Burgos
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IDISNA), CIBER-ONC CB16/12/00369, Pamplona, Spain
| | - S Böttcher
- Clinic III (Hematology, Oncology and Palliative Medicine), Special Hematology Laboratory, Rostock University Medical School, Rostock, Germany
| | - J J M van Dongen
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - A Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain; Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Neirinck J, Buysse M, De Vriendt C, Hofmans M, Bonroy C. The role of immunophenotyping in common variable immunodeficiency: a narrative review. Crit Rev Clin Lab Sci 2025; 62:65-84. [PMID: 39364936 DOI: 10.1080/10408363.2024.2404842] [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: 06/24/2024] [Revised: 08/06/2024] [Accepted: 09/12/2024] [Indexed: 10/05/2024]
Abstract
Common variable immunodeficiency (CVID) is a heterogeneous primary immunodeficiency (PID) characterized by an impaired immunoglobulin production, in association with an increased susceptibility to infections and a diversity of clinical manifestations. This narrative review summarizes immunophenotypic abnormalities in CVID patients and their relevance for diagnosis and disease classification. A comprehensive search across four databases - PubMED, Web of Science, EMBASE and Google Scholar - yielded 170 relevant studies published between 1988 and April 31, 2023. Over the past decades, the role of immunophenotyping in CVID diagnosis has become evident by identifying "hallmark" immunophenotypic aberrancies in patient subsets, with some now integrated in the consensus diagnostic criteria. Furthermore, the role of immunophenotyping in subclassifying CVID in relation to clinical presentation and prognosis has been extensively studied. Certain immunophenotypic patterns consistently correlate with clinical manifestations and/or subsets of CVID, particularly those associated with noninfectious complications (i.e. low switched memory B cells, shifts in follicular helper T cell subsets, low naïve CD4+ T cells, low regulatory T cells, and expansion of CD21low B cells, often associated with autoimmunity and/or splenomegaly). Also, efforts to associate subset levels of innate immune cells, such as Natural Killer (NK) cells, invariant (i)NKT cells, innate lymphoid cells (ILCs), and dendritic cells (DCs) to CVID complications are evident albeit in a lesser degree. However, inconsistencies regarding the role of flow cytometry in classification and prognosis persist, reflecting the disease complexity, but probably also cohort variations and methodological differences between published studies. This underscores the need for collaborative efforts to integrate emerging concepts, such as standardized flow cytometry and computational tools, for a more precise CVID classification approach. Additionally, recent studies suggest a potential value of (epi)genetic-based molecular assays to this effort.
Collapse
Affiliation(s)
- Jana Neirinck
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Malicorne Buysse
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ciel De Vriendt
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Mattias Hofmans
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
3
|
Neirinck J, Buysse M, Brdickova N, Perez-Andres M, De Vriendt C, Kerre T, Haerynck F, Bossuyt X, van Dongen JJM, Orfao A, Hofmans M, Bonroy C, Kalina T. The EuroFlow PIDOT external quality assurance scheme: enhancing laboratory performance evaluation in immunophenotyping of rare lymphoid immunodeficiencies. Clin Chem Lab Med 2025; 63:621-635. [PMID: 39423371 DOI: 10.1515/cclm-2024-0749] [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: 06/26/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES The development of External Quality Assessment Schemes (EQAS) for clinical flow cytometry (FCM) is challenging in the context of rare (immunological) diseases. Here, we introduce a novel EQAS monitoring the primary immunodeficiency Orientation Tube (PIDOT), developed by EuroFlow, in both a 'wet' and 'dry' format. This EQAS provides feedback on the quality of individual laboratories (i.e., accuracy, reproducibility and result interpretation), while eliminating the need for sample distribution. METHODS In the wet format, marker staining intensities (MedFIs) within landmark cell populations in PIDOT analysis performed on locally collected healthy control (HC) samples, were compared to EQAS targets. In the dry format, participants analyzed centrally distributed PIDOT flow cytometry data (n=10). RESULTS We report the results of six EQAS rounds across 20 laboratories in 11 countries. The wet format (212 HC samples) demonstrated consistent technical performance among laboratories (median %rCV on MedFIs=34.5 %; average failure rate 17.3 %) and showed improvement upon repeated participation. The dry format demonstrated effective proficiency of participants in cell count enumeration (range %rCVs 3.1-7.1 % for the major lymphoid subsets), and in identifying lymphoid abnormalities (79.3 % alignment with reference). CONCLUSIONS The PIDOT-EQAS allows laboratories, adhering to the standardized EuroFlow approach, to monitor interlaboratory variations without the need for sample distribution, and provides them educational support to recognize rare clinically relevant immunophenotypic patterns of primary immunodeficiencies (PID). This EQAS contributes to quality improvement of PID diagnostics and can serve as an example for future flow cytometry EQAS in the context of rare diseases.
Collapse
Affiliation(s)
- Jana Neirinck
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Malicorne Buysse
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Naděžda Brdickova
- CLIP Cytometry, Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Martín Perez-Andres
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Ciel De Vriendt
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Tessa Kerre
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Pediatric Pulmonology and Immunology and PID Research Laboratory, University Hospital Ghent, Ghent, Belgium
| | - Xavier Bossuyt
- Department of Laboratory Medicine, University Hospital Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jacques J M van Dongen
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Mattias Hofmans
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Tomas Kalina
- CLIP Cytometry, Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
4
|
Kelleher P, Greathead L, Whitby L, Brando B, Barnett D, Bloxham D, deTute R, Dunlop A, Farren T, Francis S, Payne D, Scott S, Snowden JA, Sorour Y, Stansfield E, Virgo P, Whitby A. European flow cytometry quality assurance guidelines for the diagnosis of primary immune deficiencies and assessment of immune reconstitution following B cell depletion therapies and transplantation. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:424-436. [PMID: 38940298 DOI: 10.1002/cyto.b.22195] [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: 01/15/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Over the last 15 years activity of diagnostic flow cytometry services have evolved from monitoring of CD4 T cell subsets in HIV-1 infection to screening for primary and secondary immune deficiencies syndromes and assessment of immune constitution following B cell depleting therapy and transplantation. Changes in laboratory activity in high income countries have been driven by initiation of anti-retroviral therapy (ART) in HIV-1 regardless of CD4 T cell counts, increasing recognition of primary immune deficiency syndromes and the wider application of B cell depleting therapy and transplantation in clinical practice. Laboratories should use their experience in standardization and quality assurance of CD4 T cell counting in HIV-1 infection to provide immune monitoring services to patients with primary and secondary immune deficiencies. Assessment of immune reconstitution post B cell depleting agents and transplantation can also draw on the expertise acquired by flow cytometry laboratories for detection of CD34 stem cell and assessment of MRD in hematological malignancies. This guideline provides recommendations for clinical laboratories on providing flow cytometry services in screening for immune deficiencies and its emerging role immune reconstitution after B cell targeting therapies and transplantation.
Collapse
Affiliation(s)
- Peter Kelleher
- Immunology of Infection, Department of Infectious Disease, Imperial College London, London, UK
- Department of Infection and Immunity Sciences, North West London Pathology, London, UK
| | - Louise Greathead
- Department of Infection and Immunity Sciences, North West London Pathology, London, UK
| | - Liam Whitby
- UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Bruno Brando
- Hematology Laboratory and Transfusion Center, New Hospital of Legnano: Ospedale Nuovo di Legnano, Milan, Italy
| | - David Barnett
- UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - David Bloxham
- Haematopathology and Oncology Diagnostic Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ruth deTute
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Alan Dunlop
- Department of Haemato-Oncology, Royal Marsden Hospital, London, UK
| | - Timothy Farren
- Division of Haemato-Oncology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Pathology Group, Blizard Institute, Queen Mary University of London, London, UK
| | - Sebastian Francis
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Daniel Payne
- Tees Valley Pathology Service, James Cook University Hospital, Middlesbrough, UK
| | - Stuart Scott
- UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Youssef Sorour
- Haematology, Doncaster and Bassetlaw Teaching Hospitals NHS Trust, Doncaster, UK
| | - Emma Stansfield
- Greater Manchester Immunology Service, Manchester University NHS Foundation Trust, Manchester, UK
| | - Paul Virgo
- Department of Immunology and Immunogenetics, North Bristol NHS Trust, Bristol, UK
| | - Alison Whitby
- UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
5
|
Verbeek MWC, Rodríguez BS, Sedek L, Laqua A, Buracchi C, Buysse M, Reiterová M, Oliveira E, Morf D, Oude Alink SR, Barrena S, Kohlscheen S, Nierkens S, Hofmans M, Fernandez P, de Costa ES, Mejstrikova E, Szczepanski T, Slota L, Brüggemann M, Gaipa G, Grigore G, van Dongen JJM, Orfao A, van der Velden VHJ. Minimal residual disease assessment in B-cell precursor acute lymphoblastic leukemia by semi-automated identification of normal hematopoietic cells: A EuroFlow study. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:252-263. [PMID: 37740440 DOI: 10.1002/cyto.b.22143] [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] [Received: 12/23/2022] [Revised: 07/28/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023]
Abstract
Presence of minimal residual disease (MRD), detected by flow cytometry, is an important prognostic biomarker in the management of B-cell precursor acute lymphoblastic leukemia (BCP-ALL). However, data-analysis remains mainly expert-dependent. In this study, we designed and validated an Automated Gating & Identification (AGI) tool for MRD analysis in BCP-ALL patients using the two tubes of the EuroFlow 8-color MRD panel. The accuracy, repeatability, and reproducibility of the AGI tool was validated in a multicenter study using bone marrow follow-up samples from 174 BCP-ALL patients, stained with the EuroFlow BCP-ALL MRD panel. In these patients, MRD was assessed both by manual analysis and by AGI tool supported analysis. Comparison of MRD levels obtained between both approaches showed a concordance rate of 83%, with comparable concordances between MRD tubes (tube 1, 2 or both), treatment received (chemotherapy versus targeted therapy) and flow cytometers (FACSCanto versus FACSLyric). After review of discordant cases by additional experts, the concordance increased to 97%. Furthermore, the AGI tool showed excellent intra-expert concordance (100%) and good inter-expert concordance (90%). In addition to MRD levels, also percentages of normal cell populations showed excellent concordance between manual and AGI tool analysis. We conclude that the AGI tool may facilitate MRD analysis using the EuroFlow BCP-ALL MRD protocol and will contribute to a more standardized and objective MRD assessment. However, appropriate training is required for the correct analysis of MRD data.
Collapse
Affiliation(s)
- Martijn W C Verbeek
- Laboratory for Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Beatriz Soriano Rodríguez
- Translational and Clinical Research program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Lukasz Sedek
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Zabrze, Poland
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Anna Laqua
- Department of Hematology, University of Schleswig-Holstein, Kiel, Germany
| | - Chiara Buracchi
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Malicorne Buysse
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Michaela Reiterová
- CLIP-Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Elen Oliveira
- Pediatrics Institute IPPMG, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniela Morf
- Institute for Laboratory Medicine, Aarau, Switzerland
| | - Sjoerd R Oude Alink
- Laboratory for Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Susana Barrena
- Translational and Clinical Research program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Saskia Kohlscheen
- Department of Hematology, University of Schleswig-Holstein, Kiel, Germany
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Mattias Hofmans
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | | | - Elaine Sobral de Costa
- Pediatrics Institute IPPMG, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ester Mejstrikova
- CLIP-Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Lukasz Slota
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Monika Brüggemann
- Department of Hematology, University of Schleswig-Holstein, Kiel, Germany
| | - Giuseppe Gaipa
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | | | - Jacques J M van Dongen
- Translational and Clinical Research program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Immunology, Leiden University Medical Center (LUMC), The Netherlands
| | - Alberto Orfao
- Translational and Clinical Research program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Vincent H J van der Velden
- Laboratory for Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Kowarsch F, Maurer-Granofszky M, Weijler L, Wödlinger M, Reiter M, Schumich A, Feuerstein T, Sala S, Nováková M, Faggin G, Gaipa G, Hrusak O, Buldini B, Dworzak MN. FCM marker importance for MRD assessment in T-cell acute lymphoblastic leukemia: An AIEOP-BFM-ALL-FLOW study group report. Cytometry A 2024; 105:24-35. [PMID: 37776305 DOI: 10.1002/cyto.a.24805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 08/07/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
T-lineage acute lymphoblastic leukemia (T-ALL) accounts for about 15% of pediatric and about 25% of adult ALL cases. Minimal/measurable residual disease (MRD) assessed by flow cytometry (FCM) is an important prognostic indicator for risk stratification. In order to assess the MRD a limited number of antibodies directed against the most discriminative antigens must be selected. We propose a pipeline for evaluating the influence of different markers for cell population classification in FCM data. We use linear support vector machine, fitted to each sample individually to avoid issues with patient and laboratory variations. The best separating hyperplane direction as well as the influence of omitting specific markers is considered. Ninety-one bone marrow samples of 43 pediatric T-ALL patients from five reference laboratories were analyzed by FCM regarding marker importance for blast cell identification using combinations of eight different markers. For all laboratories, CD48 and CD99 were among the top three markers with strongest contribution to the optimal hyperplane, measured by median separating hyperplane coefficient size for all samples per center and time point (diagnosis, Day 15, Day 33). Based on the available limited set tested (CD3, CD4, CD5, CD7, CD8, CD45, CD48, CD99), our findings prove that CD48 and CD99 are useful markers for MRD monitoring in T-ALL. The proposed pipeline can be applied for evaluation of other marker combinations in the future.
Collapse
Affiliation(s)
- Florian Kowarsch
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Margarita Maurer-Granofszky
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik GmbH, Vienna, Austria
| | - Lisa Weijler
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Matthias Wödlinger
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Michael Reiter
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Angela Schumich
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Tamar Feuerstein
- The Rina Zaizov Division of Pediatric Hematology-Oncology, Schneider's Children's Medical Center, Petah Tikva, Israel
| | - Simona Sala
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Michaela Nováková
- Department of Pediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Giovanni Faggin
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, Padova, Italy
| | - Giuseppe Gaipa
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Ondrej Hrusak
- Department of Pediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Barbara Buldini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, Padova, Italy
- Advanced Diagnostics and Target Discovery in ALL, Fondazione istituto di Ricerca pediatrica Città della Speranza, Padova, Italy
| | - Michael N Dworzak
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik GmbH, Vienna, Austria
| |
Collapse
|
7
|
Delgado AH, Fluxa R, Perez-Andres M, Diks AM, van Gaans-van den Brink JAM, Barkoff AM, Blanco E, Torres-Valle A, Berkowska MA, Grigore G, van Dongen J.J.M, Orfao A. Automated EuroFlow approach for standardized in-depth dissection of human circulating B-cells and plasma cells. Front Immunol 2023; 14:1268686. [PMID: 37915569 PMCID: PMC10616957 DOI: 10.3389/fimmu.2023.1268686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Background Multiparameter flow cytometry (FC) immunophenotyping is a key tool for detailed identification and characterization of human blood leucocytes, including B-lymphocytes and plasma cells (PC). However, currently used conventional data analysis strategies require extensive expertise, are time consuming, and show limited reproducibility. Objective Here, we designed, constructed and validated an automated database-guided gating and identification (AGI) approach for fast and standardized in-depth dissection of B-lymphocyte and PC populations in human blood. Methods For this purpose, 213 FC standard (FCS) datafiles corresponding to umbilical cord and peripheral blood samples from healthy and patient volunteers, stained with the 14-color 18-antibody EuroFlow BIgH-IMM panel, were used. Results The BIgH-IMM antibody panel allowed identification of 117 different B-lymphocyte and PC subsets. Samples from 36 healthy donors were stained and 14 of the datafiles that fulfilled strict inclusion criteria were analysed by an expert flow cytometrist to build the EuroFlow BIgH-IMM database. Data contained in the datafiles was then merged into a reference database that was uploaded in the Infinicyt software (Cytognos, Salamanca, Spain). Subsequently, we compared the results of manual gating (MG) with the performance of two classification algorithms -hierarchical algorithm vs two-step algorithm- for AGI of the cell populations present in 5 randomly selected FCS datafiles. The hierarchical AGI algorithm showed higher correlation values vs conventional MG (r2 of 0.94 vs. 0.88 for the two-step AGI algorithm) and was further validated in a set of 177 FCS datafiles against conventional expert-based MG. For virtually all identifiable cell populations a highly significant correlation was observed between the two approaches (r2>0.81 for 79% of all B-cell populations identified), with a significantly lower median time of analysis per sample (6 vs. 40 min, p=0.001) for the AGI tool vs. MG, respectively and both intra-sample (median CV of 1.7% vs. 10.4% by MG, p<0.001) and inter-expert (median CV of 3.9% vs. 17.3% by MG by 2 experts, p<0.001) variability. Conclusion Our results show that compared to conventional FC data analysis strategies, the here proposed AGI tool is a faster, more robust, reproducible, and standardized approach for in-depth analysis of B-lymphocyte and PC subsets circulating in human blood.
Collapse
Affiliation(s)
- Alejandro H. Delgado
- Cytognos SL, Salamanca, Spain
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
| | | | - Martin Perez-Andres
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Annieck M. Diks
- Department of Immunology (IMMU), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | | | - Elena Blanco
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Alba Torres-Valle
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Magdalena A. Berkowska
- Department of Immunology (IMMU), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | - J .J .M. van Dongen
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Immunology (IMMU), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Medina-Herrera A, Sarasquete ME, Jiménez C, Puig N, García-Sanz R. Minimal Residual Disease in Multiple Myeloma: Past, Present, and Future. Cancers (Basel) 2023; 15:3687. [PMID: 37509348 PMCID: PMC10377959 DOI: 10.3390/cancers15143687] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Responses to treatment have improved over the last decades for patients with multiple myeloma. This is a consequence of the introduction of new drugs that have been successfully combined in different clinical contexts: newly diagnosed, transplant-eligible or ineligible patients, as well as in the relapsed/refractory setting. However, a great proportion of patients continue to relapse, even those achieving complete response, which underlines the need for updated response criteria. In 2014, the international myeloma working group established new levels of response, prompting the evaluation of minimal residual disease (MRD) for those patients already in complete or stringent complete response as defined by conventional serological assessments: the absence of tumor plasma cells in 100,000 total cells or more define molecular and immunophenotypic responses by next-generation sequencing and flow cytometry, respectively. In this review, we describe all the potential methods that may be used for MRD detection based on the evidence found in the literature, paying special attention to their advantages and pitfalls from a critical perspective.
Collapse
Affiliation(s)
- Alejandro Medina-Herrera
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - María Eugenia Sarasquete
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Cristina Jiménez
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Noemí Puig
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Ramón García-Sanz
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| |
Collapse
|
9
|
Roca CP, Burton OT, Neumann J, Tareen S, Whyte CE, Gergelits V, Veiga RV, Humblet-Baron S, Liston A. A cross entropy test allows quantitative statistical comparison of t-SNE and UMAP representations. CELL REPORTS METHODS 2023; 3:100390. [PMID: 36814837 PMCID: PMC9939422 DOI: 10.1016/j.crmeth.2022.100390] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/29/2022] [Accepted: 12/20/2022] [Indexed: 01/15/2023]
Abstract
The advent of high-dimensional single-cell data has necessitated the development of dimensionality-reduction tools. t-Distributed stochastic neighbor embedding (t-SNE) and uniform manifold approximation and projection (UMAP) are the two most frequently used approaches, allowing clear visualization of complex single-cell datasets. Despite the need for quantitative comparison, t-SNE and UMAP have largely remained visualization tools due to the lack of robust statistical approaches. Here, we have derived a statistical test for evaluating the difference between dimensionality-reduced datasets using the Kolmogorov-Smirnov test on the distributions of cross entropy of single cells within each dataset. As the approach uses the inter-relationship of single cells for comparison, the resulting statistic is robust and capable of identifying true biological variation. Further, the test provides a valid distance between single-cell datasets, allowing the organization of multiple samples into a dendrogram for quantitative comparison of complex datasets. These results demonstrate the largely untapped potential of dimensionality-reduction tools for biomedical data analysis beyond visualization.
Collapse
Affiliation(s)
- Carlos P. Roca
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Oliver T. Burton
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Julika Neumann
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, 3000 Leuven, Belgium
| | - Samar Tareen
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Carly E. Whyte
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Vaclav Gergelits
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Rafael V. Veiga
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | | - Adrian Liston
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| |
Collapse
|
10
|
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
|
11
|
Lemieux ME, Reveles XT, Rebeles J, Bederka LH, Araujo PR, Sanchez JR, Grayson M, Lai SC, DePalo LR, Habib SA, Hill DG, Lopez K, Patriquin L, Sussman R, Joyce RP, Rebel VI. Detection of early-stage lung cancer in sputum using automated flow cytometry and machine learning. Respir Res 2023; 24:23. [PMID: 36681813 PMCID: PMC9862555 DOI: 10.1186/s12931-023-02327-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Low-dose spiral computed tomography (LDCT) may not lead to a clear treatment path when small to intermediate-sized lung nodules are identified. We have combined flow cytometry and machine learning to develop a sputum-based test (CyPath Lung) that can assist physicians in decision-making in such cases. METHODS Single cell suspensions prepared from induced sputum samples collected over three consecutive days were labeled with a viability dye to exclude dead cells, antibodies to distinguish cell types, and a porphyrin to label cancer-associated cells. The labeled cell suspension was run on a flow cytometer and the data collected. An analysis pipeline combining automated flow cytometry data processing with machine learning was developed to distinguish cancer from non-cancer samples from 150 patients at high risk of whom 28 had lung cancer. Flow data and patient features were evaluated to identify predictors of lung cancer. Random training and test sets were chosen to evaluate predictive variables iteratively until a robust model was identified. The final model was tested on a second, independent group of 32 samples, including six samples from patients diagnosed with lung cancer. RESULTS Automated analysis combined with machine learning resulted in a predictive model that achieved an area under the ROC curve (AUC) of 0.89 (95% CI 0.83-0.89). The sensitivity and specificity were 82% and 88%, respectively, and the negative and positive predictive values 96% and 61%, respectively. Importantly, the test was 92% sensitive and 87% specific in cases when nodules were < 20 mm (AUC of 0.94; 95% CI 0.89-0.99). Testing of the model on an independent second set of samples showed an AUC of 0.85 (95% CI 0.71-0.98) with an 83% sensitivity, 77% specificity, 95% negative predictive value and 45% positive predictive value. The model is robust to differences in sample processing and disease state. CONCLUSION CyPath Lung correctly classifies samples as cancer or non-cancer with high accuracy, including from participants at different disease stages and with nodules < 20 mm in diameter. This test is intended for use after lung cancer screening to improve early-stage lung cancer diagnosis. Trial registration ClinicalTrials.gov ID: NCT03457415; March 7, 2018.
Collapse
Affiliation(s)
| | - Xavier T. Reveles
- bioAffinity Technologies, 22211 W I-10, Suite 1206, San Antonio, TX 78257 USA
| | - Jennifer Rebeles
- bioAffinity Technologies, 22211 W I-10, Suite 1206, San Antonio, TX 78257 USA
| | - Lydia H. Bederka
- bioAffinity Technologies, 22211 W I-10, Suite 1206, San Antonio, TX 78257 USA
| | - Patricia R. Araujo
- bioAffinity Technologies, 22211 W I-10, Suite 1206, San Antonio, TX 78257 USA
| | - Jamila R. Sanchez
- bioAffinity Technologies, 22211 W I-10, Suite 1206, San Antonio, TX 78257 USA
| | - Marcia Grayson
- bioAffinity Technologies, 22211 W I-10, Suite 1206, San Antonio, TX 78257 USA
| | - Shao-Chiang Lai
- bioAffinity Technologies, 22211 W I-10, Suite 1206, San Antonio, TX 78257 USA
| | - Louis R. DePalo
- grid.59734.3c0000 0001 0670 2351Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Sheila A. Habib
- grid.414059.d0000 0004 0617 9080South Texas Veterans Health Care System (STVHCS), Audie L. Murphy Memorial Veterans Hospital, San Antonio, TX USA
| | - David G. Hill
- Waterbury Pulmonary Associates LLC, Waterbury, CT USA
| | - Kathleen Lopez
- grid.477754.2Radiology Associates of Albuquerque, Albuquerque, NM USA
| | - Lara Patriquin
- grid.477754.2Radiology Associates of Albuquerque, Albuquerque, NM USA ,Present Address: Zia Diagnostic Imaging, Albuquerque, NM USA
| | | | | | - Vivienne I. Rebel
- bioAffinity Technologies, 22211 W I-10, Suite 1206, San Antonio, TX 78257 USA
| |
Collapse
|
12
|
van der Pan K, de Bruin-Versteeg S, Damasceno D, Hernández-Delgado A, van der Sluijs-Gelling AJ, van den Bossche WBL, de Laat IF, Díez P, Naber BAE, Diks AM, Berkowska MA, de Mooij B, Groenland RJ, de Bie FJ, Khatri I, Kassem S, de Jager AL, Louis A, Almeida J, van Gaans-van den Brink JAM, Barkoff AM, He Q, Ferwerda G, Versteegen P, Berbers GAM, Orfao A, van Dongen JJM, Teodosio C. Development of a standardized and validated flow cytometry approach for monitoring of innate myeloid immune cells in human blood. Front Immunol 2022; 13:935879. [PMID: 36189252 PMCID: PMC9519388 DOI: 10.3389/fimmu.2022.935879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Innate myeloid cell (IMC) populations form an essential part of innate immunity. Flow cytometric (FCM) monitoring of IMCs in peripheral blood (PB) has great clinical potential for disease monitoring due to their role in maintenance of tissue homeostasis and ability to sense micro-environmental changes, such as inflammatory processes and tissue damage. However, the lack of standardized and validated approaches has hampered broad clinical implementation. For accurate identification and separation of IMC populations, 62 antibodies against 44 different proteins were evaluated. In multiple rounds of EuroFlow-based design-testing-evaluation-redesign, finally 16 antibodies were selected for their non-redundancy and separation power. Accordingly, two antibody combinations were designed for fast, sensitive, and reproducible FCM monitoring of IMC populations in PB in clinical settings (11-color; 13 antibodies) and translational research (14-color; 16 antibodies). Performance of pre-analytical and analytical variables among different instruments, together with optimized post-analytical data analysis and reference values were assessed. Overall, 265 blood samples were used for design and validation of the antibody combinations and in vitro functional assays, as well as for assessing the impact of sample preparation procedures and conditions. The two (11- and 14-color) antibody combinations allowed for robust and sensitive detection of 19 and 23 IMC populations, respectively. Highly reproducible identification and enumeration of IMC populations was achieved, independently of anticoagulant, type of FCM instrument and center, particularly when database/software-guided automated (vs. manual “expert-based”) gating was used. Whereas no significant changes were observed in identification of IMC populations for up to 24h delayed sample processing, a significant impact was observed in their absolute counts after >12h delay. Therefore, accurate identification and quantitation of IMC populations requires sample processing on the same day. Significantly different counts were observed in PB for multiple IMC populations according to age and sex. Consequently, PB samples from 116 healthy donors (8-69 years) were used for collecting age and sex related reference values for all IMC populations. In summary, the two antibody combinations and FCM approach allow for rapid, standardized, automated and reproducible identification of 19 and 23 IMC populations in PB, suited for monitoring of innate immune responses in clinical and translational research settings.
Collapse
Affiliation(s)
- Kyra van der Pan
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Daniela Damasceno
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Alejandro Hernández-Delgado
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | | | - Wouter B. L. van den Bossche
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Department of Immunology, Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Inge F. de Laat
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Paula Díez
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Annieck M. Diks
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Bas de Mooij
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Rick J. Groenland
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Fenna J. de Bie
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Indu Khatri
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Sara Kassem
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Anniek L. de Jager
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Alesha Louis
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | | | - Alex-Mikael Barkoff
- Institute of Biomedicine, Research Center for Infections and Immunity, University of Turku (UTU), Turku, Finland
| | - Qiushui He
- Institute of Biomedicine, Research Center for Infections and Immunity, University of Turku (UTU), Turku, Finland
| | - Gerben Ferwerda
- Section of Paediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Pauline Versteegen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Guy A. M. Berbers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Jacques J. M. van Dongen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- *Correspondence: Jacques J. M. van Dongen,
| | - Cristina Teodosio
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
13
|
Neirinck J, Emmaneel A, Buysse M, Philippé J, Van Gassen S, Saeys Y, Bossuyt X, De Buyser S, van der Burg M, Pérez-Andrés M, Orfao A, van Dongen JJM, Lambrecht BN, Kerre T, Hofmans M, Haerynck F, Bonroy C. The Euroflow PID Orientation Tube in the diagnostic workup of primary immunodeficiency: Daily practice performance in a tertiary university hospital. Front Immunol 2022; 13:937738. [PMID: 36177024 PMCID: PMC9513319 DOI: 10.3389/fimmu.2022.937738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Multiparameter flow cytometry (FCM) immunophenotyping is an important tool in the diagnostic screening and classification of primary immunodeficiencies (PIDs). The EuroFlow Consortium recently developed the PID Orientation Tube (PIDOT) as a universal screening tool to identify lymphoid-PID in suspicious patients. Although PIDOT can identify different lymphoid-PIDs with high sensitivity, clinical validation in a broad spectrum of patients with suspicion of PID is missing. In this study, we investigated the diagnostic performance of PIDOT, as part of the EuroFlow diagnostic screening algorithm for lymphoid-PID, in a daily practice at a tertiary reference center for PID. Methods PIDOT was tested in 887 consecutive patients suspicious of PID at the Ghent University Hospital, Belgium. Patients were classified into distinct subgroups of lymphoid-PID vs. non-PID disease controls (non-PID DCs), according to the IUIS and ESID criteria. For the clinical validation of PIDOT, comprehensive characterization of the lymphoid defects was performed, together with the identification of the most discriminative cell subsets to distinguish lymphoid-PID from non-PID DCs. Next, a decision-tree algorithm was designed to guide subsequent FCM analyses. Results The mean number of lymphoid defects detected by PIDOT in blood was 2.87 times higher in lymphoid-PID patients vs. non-PID DCs (p < 0.001), resulting in an overall sensitivity and specificity of 87% and 62% to detect severe combined immunodeficiency (SCID), combined immunodeficiency with associated or syndromic features (CID), immune dysregulation disorder (ID), and common variable immunodeficiency (CVID). The most discriminative populations were total memory and switched memory B cells, total T cells, TCD4+cells, and naive TCD4+cells, together with serum immunoglobulin levels. Based on these findings, a decision-tree algorithm was designed to guide further FCM analyses, which resulted in an overall sensitivity and specificity for all lymphoid-PIDs of 86% and 82%, respectively. Conclusion Altogether, our findings confirm that PIDOT is a powerful tool for the diagnostic screening of lymphoid-PID, particularly to discriminate (S)CID, ID, and CVID patients from other patients suspicious of PID. The combination of PIDOT and serum immunoglobulin levels provides an efficient guide for further immunophenotypic FCM analyses, complementary to functional and genetic assays, for accurate PID diagnostics.
Collapse
Affiliation(s)
- Jana Neirinck
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Annelies Emmaneel
- Data Mining and Modelling for Biomedicine Group, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Malicorne Buysse
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jan Philippé
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Sofie Van Gassen
- Data Mining and Modelling for Biomedicine Group, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Yvan Saeys
- Data Mining and Modelling for Biomedicine Group, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Xavier Bossuyt
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, KU Leuven University Hospitals Leuven, Leuven, Belgium
| | - Stefanie De Buyser
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Mirjam van der Burg
- Laboratory for Pediatric Immunology, Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Martín Pérez-Andrés
- Cancer Research Centre (Instituto de Biología Molecular y Celular del Cáncer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), University of Salamanca, Salamanca, Spain
| | - Alberto Orfao
- Cancer Research Centre (Instituto de Biología Molecular y Celular del Cáncer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), University of Salamanca, Salamanca, Spain
| | | | - Bart N. Lambrecht
- Laboratory of Mucosal Immunology, VIB-UGhent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, University Hospital Ghent, Ghent, Belgium
| | - Tessa Kerre
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Mattias Hofmans
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Pediatric Pulmonology and Immunology and Primary Immunodeficiency (PID) Research Lab, Ghent University Hospital, Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
- *Correspondence: Carolien Bonroy,
| |
Collapse
|
14
|
Couckuyt A, Seurinck R, Emmaneel A, Quintelier K, Novak D, Van Gassen S, Saeys Y. Challenges in translational machine learning. Hum Genet 2022; 141:1451-1466. [PMID: 35246744 PMCID: PMC8896412 DOI: 10.1007/s00439-022-02439-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 02/08/2022] [Indexed: 11/25/2022]
Abstract
Machine learning (ML) algorithms are increasingly being used to help implement clinical decision support systems. In this new field, we define as "translational machine learning", joint efforts and strong communication between data scientists and clinicians help to span the gap between ML and its adoption in the clinic. These collaborations also improve interpretability and trust in translational ML methods and ultimately aim to result in generalizable and reproducible models. To help clinicians and bioinformaticians refine their translational ML pipelines, we review the steps from model building to the use of ML in the clinic. We discuss experimental setup, computational analysis, interpretability and reproducibility, and emphasize the challenges involved. We highly advise collaboration and data sharing between consortia and institutes to build multi-centric cohorts that facilitate ML methodologies that generalize across centers. In the end, we hope that this review provides a way to streamline translational ML and helps to tackle the challenges that come with it.
Collapse
Affiliation(s)
- Artuur Couckuyt
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Gent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Gent, Belgium
| | - Ruth Seurinck
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Gent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Gent, Belgium
| | - Annelies Emmaneel
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Gent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Gent, Belgium
| | - Katrien Quintelier
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Gent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Gent, Belgium
- Department of Pulmonary Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - David Novak
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Gent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Gent, Belgium
| | - Sofie Van Gassen
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Gent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Gent, Belgium
| | - Yvan Saeys
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Gent, Belgium.
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Gent, Belgium.
| |
Collapse
|
15
|
Bras AE, Matarraz S, Nierkens S, Fernández P, Philippé J, Aanei CM, de Mello FV, Burgos L, van der Sluijs-Gelling AJ, Grigore GE, van Dongen JJM, Orfao A, van der Velden VHJ. Quality Assessment of a Large Multi-Center Flow Cytometric Dataset of Acute Myeloid Leukemia Patients-A EuroFlow Study. Cancers (Basel) 2022; 14:cancers14082011. [PMID: 35454917 PMCID: PMC9033003 DOI: 10.3390/cancers14082011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
Flowcytometric analysis allows for detailed identification and characterization of large numbers of cells in blood, bone marrow, and other body fluids and tissue samples and therefore contributes to the diagnostics of hematological malignancies. Novel data analysis tools allow for multidimensional analysis and comparison of patient samples with reference databases of normal, reactive, and/or leukemia/lymphoma patient samples. Building such reference databases requires strict quality assessment (QA) procedures. Here, we compiled a dataset and developed a QA methodology of the EuroFlow Acute Myeloid Leukemia (AML) database, based on the eight-color EuroFlow AML panel consisting of six different antibody combinations, including four backbone markers. In total, 1142 AML cases and 42 normal bone marrow samples were included in this analysis. QA was performed on 803 AML cases using multidimensional analysis of backbone markers, as well as tube-specific markers, and data were compared using classical analysis employing median and peak expression values. Validation of the QA procedure was performed by re-analysis of >300 cases and by running an independent cohort of 339 AML cases. Initial evaluation of the final cohort confirmed specific immunophenotypic patterns in AML subgroups; the dataset therefore can reliably be used for more detailed exploration of the immunophenotypic variability of AML. Our data show the potential pitfalls and provide possible solutions for constructing large flowcytometric databases. In addition, the provided approach may facilitate the building of other databases and thereby support the development of novel tools for (semi)automated QA and subsequent data analysis.
Collapse
Affiliation(s)
- Anne E. Bras
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands;
| | - Sergio Matarraz
- Cancer Research Center (IBMCC-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, CIBERONC and Institute of Biomedical Research of Salamanca (IBSAL), Campus Miguel de Unamuno, Paseo de la Universidad de Coimbra s/n, 37007 Salamanca, Spain; (S.M.); (A.O.)
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands;
| | - Paula Fernández
- Institute for Laboratory Medicine, Kantonsspital Aarau AG, Tellstrasse 25, 5001 Aarau, Switzerland;
| | - Jan Philippé
- Department of Diagnostic Sciences, Ghent University, C. Heymanslaan 10, 9000 Ghent, Belgium;
| | - Carmen-Mariana Aanei
- Laboratory of Hematology, University Hospital of Saint-Etienne, Av. Albert Raimond, 42055 Saint-Etienne, France;
| | - Fabiana Vieira de Mello
- Cytometry Service, Institute of Pediatrics (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rua Bruno Lobo 50, Cidade Universitária, Rio de Janeiro 21941-912, RJ, Brazil;
| | - Leire Burgos
- Applied Medical Research Center (CIMA), Instituto de Investigacion Sanitaria de Navarra (IDISNA), Clinica Universidad de Navarra, 31008 Pamplona, Spain;
| | - Alita J. van der Sluijs-Gelling
- Department of Immunology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (A.J.v.d.S.-G.); (J.J.M.v.D.)
| | - Georgiana Emilia Grigore
- Cytognos SL, Carretera de Madrid Km. 0 Nave 9, Pol. La Serna, Santa Marta de Tormes, 37900 Salamanca, Spain;
| | - Jacques J. M. van Dongen
- Department of Immunology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (A.J.v.d.S.-G.); (J.J.M.v.D.)
- Cancer Research Center (CIC), Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Alberto Orfao
- Cancer Research Center (IBMCC-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, CIBERONC and Institute of Biomedical Research of Salamanca (IBSAL), Campus Miguel de Unamuno, Paseo de la Universidad de Coimbra s/n, 37007 Salamanca, Spain; (S.M.); (A.O.)
| | - Vincent H. J. van der Velden
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands;
- Correspondence: ; Tel.: +31-10-704-4253
| | | |
Collapse
|
16
|
Monaghan SA, Li JL, Liu YC, Ko MY, Boyiadzis M, Chang TY, Wang YF, Lee CC, Swerdlow SH, Ko BS. A Machine Learning Approach to the Classification of Acute Leukemias and Distinction From Nonneoplastic Cytopenias Using Flow Cytometry Data. Am J Clin Pathol 2022; 157:546-553. [PMID: 34643210 DOI: 10.1093/ajcp/aqab148] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/01/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Flow cytometry (FC) is critical for the diagnosis and monitoring of hematologic malignancies. Machine learning (ML) methods rapidly classify multidimensional data and should dramatically improve the efficiency of FC data analysis. We aimed to build a model to classify acute leukemias, including acute promyelocytic leukemia (APL), and distinguish them from nonneoplastic cytopenias. We also sought to illustrate a method to identify key FC parameters that contribute to the model's performance. METHODS Using data from 531 patients who underwent evaluation for cytopenias and/or acute leukemia, we developed an ML model to rapidly distinguish among APL, acute myeloid leukemia/not APL, acute lymphoblastic leukemia, and nonneoplastic cytopenias. Unsupervised learning using gaussian mixture model and Fisher kernel methods were applied to FC listmode data, followed by supervised support vector machine classification. RESULTS High accuracy (ACC, 94.2%; area under the curve [AUC], 99.5%) was achieved based on the 37-parameter FC panel. Using only 3 parameters, however, yielded similar performance (ACC, 91.7%; AUC, 98.3%) and highlighted the significant contribution of light scatter properties. CONCLUSIONS Our findings underscore the potential for ML to automatically identify and prioritize FC specimens that have critical results, including APL and other acute leukemias.
Collapse
Affiliation(s)
- Sara A Monaghan
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Presbyterian, Pittsburgh, PA, USA
| | - Jeng-Lin Li
- Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yen-Chun Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ming-Ya Ko
- Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Michael Boyiadzis
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | | | - Chi-Chun Lee
- Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Steven H Swerdlow
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Presbyterian, Pittsburgh, PA, USA
| | - Bor-Sheng Ko
- Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
17
|
Brestoff JR, Frater JL. Contemporary Challenges in Clinical Flow Cytometry: Small Samples, Big Data, Little Time. J Appl Lab Med 2022; 7:931-944. [DOI: 10.1093/jalm/jfab176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022]
Abstract
Abstract
Background
Immunophenotypic analysis of cell populations by flow cytometry has an established role in primary diagnosis and disease monitoring of many hematologic diseases. A persistent problem in evaluation of specimens is suboptimal cell counts and low cell viability, which results in an undesirable rate of analysis failure. In addition, the increased amount of data generated in flow cytometry challenges existing data analysis and reporting paradigms.
Content
We describe current and emerging technological improvements in cell analysis that allow the clinical laboratory to perform multiparameter analysis of specimens, including those with low cell counts and other quality issues. These technologies include conventional multicolor flow cytometry and new high-dimensional technologies, such as spectral flow cytometry and mass cytometry that enable detection of over 40 antigens simultaneously. The advantages and disadvantages of each approach are discussed. We also describe new innovations in flow cytometry data analysis, including artificial intelligence-aided techniques.
Summary
Improvements in analytical technology, in tandem with innovations in data analysis, data storage, and reporting mechanisms, help to optimize the quality of clinical flow cytometry. These improvements are essential because of the expanding role of flow cytometry in patient care.
Collapse
Affiliation(s)
- Jonathan R Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - John L Frater
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
18
|
Age and Primary Vaccination Background Influence the Plasma Cell Response to Pertussis Booster Vaccination. Vaccines (Basel) 2022; 10:vaccines10020136. [PMID: 35214595 PMCID: PMC8878388 DOI: 10.3390/vaccines10020136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023] Open
Abstract
Pertussis is a vaccine-preventable disease caused by the bacterium Bordetella pertussis. Over the past years, the incidence and mortality of pertussis increased significantly. A possible cause is the switch from whole-cell to acellular pertussis vaccines, although other factors may also contribute. Here, we applied high-dimensional flow cytometry to investigate changes in B cells in individuals of different ages and distinct priming backgrounds upon administration of an acellular pertussis booster vaccine. Participants were divided over four age cohorts. We compared longitudinal kinetics within each cohort and between the different cohorts. Changes in the B-cell compartment were correlated to numbers of vaccine-specific B- and plasma cells and serum Ig levels. Expansion and maturation of plasma cells 7 days postvaccination was the most prominent cellular change in all age groups and was most pronounced for more mature IgG1+ plasma cells. Plasma cell responses were stronger in individuals primed with whole-cell vaccine than in individuals primed with acellular vaccine. Moreover, IgG1+ and IgA1+ plasma cell expansion correlated with FHA-, Prn-, or PT- specific serum IgG or IgA levels. Our study indicates plasma cells as a potential early cellular marker of an immune response and contributes to understanding differences in immune responses between age groups and primary vaccination backgrounds.
Collapse
|
19
|
Aanei CM, Veyrat-Masson R, Selicean C, Marian M, Rigollet L, Trifa AP, Tomuleasa C, Serban A, Cherry M, Flandrin-Gresta P, Tardy ET, Guyotat D, Campos Catafal L. Database-Guided Analysis for Immunophenotypic Diagnosis and Follow-Up of Acute Myeloid Leukemia With Recurrent Genetic Abnormalities. Front Oncol 2021; 11:746951. [PMID: 34804933 PMCID: PMC8602100 DOI: 10.3389/fonc.2021.746951] [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: 07/25/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
Acute myeloid leukemias (AMLs) are hematologic malignancies with varied molecular and immunophenotypic profiles, making them difficult to diagnose and classify. High-dimensional analysis algorithms might increase the utility of multicolor flow cytometry for AML diagnosis and follow-up. The objective of the present study was to assess whether a Compass database-guided analysis can be used to achieve rapid and accurate diagnoses. We conducted this study to determine whether this method could be employed to pilote the genetic and molecular tests and to objectively identify different-from-normal (DfN) patterns to improve measurable residual disease follow-up in AML. Three Compass databases were built using Infinicyt 2.0 software, including normal myeloid-committed hematopoietic precursors (n = 20) and AML blasts harboring the most frequent recurrent genetic abnormalities (n = 50). The diagnostic accuracy of the Compass database-guided analysis was evaluated in a prospective validation study (125 suspected AML patients). This method excluded AML associated with the following genetic abnormalities: t(8;21), t(15;17), inv(16), and KMT2A translocation, with 92% sensitivity [95% confidence interval (CI): 78.6%–98.3%] and a 98.5% negative predictive value (95% CI: 90.6%–99.8%). Our data showed that the Compass database-guided analysis could identify phenotypic differences between AML groups, representing a useful tool for the identification of DfN patterns.
Collapse
Affiliation(s)
- Carmen-Mariana Aanei
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | - Richard Veyrat-Masson
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Cristina Selicean
- Department of Hematology, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Laboratory of Hematology, Oncological Institute "Prof. Dr. Ion Chiricuță", Cluj-Napoca, Romania
| | - Mirela Marian
- Laboratory of Hematology, Oncological Institute "Prof. Dr. Ion Chiricuță", Cluj-Napoca, Romania
| | - Lauren Rigollet
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | - Adrian Pavel Trifa
- Department of Medical Genetics, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Genetics, Oncological Institute "Prof. Dr. Ion Chiricuță", Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Clinical Hematology, Oncological Institute "Prof. Dr. Ion Chiricuță", Cluj-Napoca, Romania
| | - Adrian Serban
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | - Mohamad Cherry
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | - Pascale Flandrin-Gresta
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | - Emmanuelle Tavernier Tardy
- Département d'Hématologie Clinique, Institut de Cancérologie Lucien Neuwirth, Saint-Priest-en-Jarez, France
| | - Denis Guyotat
- Département d'Hématologie Clinique, Institut de Cancérologie Lucien Neuwirth, Saint-Priest-en-Jarez, France
| | - Lydia Campos Catafal
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
20
|
Aanei CM, Veyrat-Masson R, Rigollet L, Stagnara J, Tavernier Tardy E, Daguenet E, Guyotat D, Campos Catafal L. Advanced Flow Cytometry Analysis Algorithms for Optimizing the Detection of "Different From Normal" Immunophenotypes in Acute Myeloid Blasts. Front Cell Dev Biol 2021; 9:735518. [PMID: 34650981 PMCID: PMC8506133 DOI: 10.3389/fcell.2021.735518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukemias (AMLs) are a group of hematologic malignancies that are heterogeneous in their molecular and immunophenotypic profiles. Identification of the immunophenotypic differences between AML blasts and normal myeloid hematopoietic precursors (myHPCs) is a prerequisite to achieving better performance in AML measurable residual disease follow-ups. In the present study, we applied high-dimensional analysis algorithms provided by the Infinicyt 2.0 and Cytobank software to evaluate the efficacy of antibody combinations of the EuroFlow AML/myelodysplastic syndrome panel to distinguish AML blasts with recurrent genetic abnormalities (n = 39 AML samples) from normal CD45low CD117+ myHPCs (n = 23 normal bone marrow samples). Two types of scores were established to evaluate the abilities of the various methods to identify the most useful parameters/markers for distinguishing between AML blasts and normal myHPCs, as well as to distinguish between different AML groups. The Infinicyt Compass database-guided analysis was found to be a more user-friendly tool than other analysis methods implemented in the Cytobank software. According to the developed scoring systems, the principal component analysis based algorithms resulted in better discrimination between AML blasts and myHPCs, as well as between blasts from different AML groups. The most informative markers for the discrimination between myHPCs and AML blasts were CD34, CD36, human leukocyte antigen-DR (HLA-DR), CD13, CD105, CD71, and SSC, which were highly rated by all evaluated analysis algorithms. The HLA-DR, CD34, CD13, CD64, CD33, CD117, CD71, CD36, CD11b, SSC, and FSC were found to be useful for the distinction between blasts from different AML groups associated with recurrent genetic abnormalities. This study identified both benefits and the drawbacks of integrating multiple high-dimensional algorithms to gain complementary insights into the flow-cytometry data.
Collapse
Affiliation(s)
- Carmen-Mariana Aanei
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| | - Richard Veyrat-Masson
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Lauren Rigollet
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| | - Jérémie Stagnara
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| | | | | | - Denis Guyotat
- Institut de Cancérologie Lucien Neuwirth, Saint-Priest-en-Jarez, France
| | - Lydia Campos Catafal
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| |
Collapse
|
21
|
Del Pino-Molina L, López-Granados E, Lecrevisse Q, Torres Canizales J, Pérez-Andrés M, Blanco E, Wentink M, Bonroy C, Nechvatalova J, Milota T, Kienzler AK, Philippé J, Sousa AE, van der Burg M, Kalina T, van Dongen JJM, Orfao A. Dissection of the Pre-Germinal Center B-Cell Maturation Pathway in Common Variable Immunodeficiency Based on Standardized Flow Cytometric EuroFlow Tools. Front Immunol 2021; 11:603972. [PMID: 33679693 PMCID: PMC7925888 DOI: 10.3389/fimmu.2020.603972] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/29/2020] [Indexed: 12/03/2022] Open
Abstract
Introduction Common Variable Immunodeficiency (CVID) is characterized by defective antibody production and hypogammaglobulinemia. Flow cytometry immunophenotyping of blood lymphocytes has become of great relevance for the diagnosis and classification of CVID, due to an impaired differentiation of mature post-germinal-center (GC) class-switched memory B-cells (MBC) and severely decreased plasmablast/plasma cell (Pb) counts. Here, we investigated in detail the pre-GC B-cell maturation compartment in blood of CVID patients. Methods In this collaborative multicentric study the EuroFlow PID 8-color Pre-GC B-cell tube, standardized sample preparation procedures (SOPs) and innovative data analysis tools, were used to characterize the maturation profile of pre-GC B-cells in 100 CVID patients, vs 62 age-matched healthy donors (HD). Results The Pre-GC B-cell tube allowed identification within pre-GC B-cells of three subsets of maturation associated immature B-cells and three subpopulations of mature naïve B-lymphocytes. CVID patients showed overall reduced median absolute counts (vs HD) of the two more advanced stages of maturation of both CD5+ CD38+/++ CD21het CD24++ (2.7 vs 5.6 cells/µl, p=0.0004) and CD5+ CD38het CD21+ CD24+ (6.5 vs 17 cells/µl, p<0.0001) immature B cells (below normal HD levels in 22% and 37% of CVID patients). This was associated with an expansion of CD21-CD24- (6.1 vs 0.74 cells/µl, p<0.0001) and CD21-CD24++ (1.8 vs 0.4 cells/µl, p<0.0001) naïve B-cell counts above normal values in 73% and 94% cases, respectively. Additionally, reduced IgMD+ (21 vs 32 cells/µl, p=0.03) and IgMD- (4 vs 35 cells/µl, p<0.0001) MBC counts were found to be below normal values in 25% and 77% of CVID patients, respectively, always together with severely reduced/undetectable circulating blood pb. Comparison of the maturation pathway profile of pre-GC B cells in blood of CVID patients vs HD using EuroFlow software tools showed systematically altered patterns in CVID. These consisted of: i) a normally-appearing maturation pathway with altered levels of expression of >1 (CD38, CD5, CD19, CD21, CD24, and/or smIgM) phenotypic marker (57/88 patients; 65%) for a total of 3 distinct CVID patient profiles (group 1: 42/88 patients, 48%; group 2: 8/88, 9%; and group 3: 7/88, 8%) and ii) CVID patients with a clearly altered pre-GC B cell maturation pathway in blood (group 4: 31/88 cases, 35%). Conclusion Our results show that maturation of pre-GC B-cells in blood of CVID is systematically altered with up to four distinctly altered maturation profiles. Further studies, are necessary to better understand the impact of such alterations on the post-GC defects and the clinical heterogeneity of CVID.
Collapse
Affiliation(s)
- Lucía Del Pino-Molina
- Clinical Immunology Department, La Paz University Hospital and Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ) and Center for Biomedical Network Research on Rare Diseases (CIBERER U767), Madrid, Spain
| | - Eduardo López-Granados
- Clinical Immunology Department, La Paz University Hospital and Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ) and Center for Biomedical Network Research on Rare Diseases (CIBERER U767), Madrid, Spain
| | - Quentin Lecrevisse
- Clinical and Translation Research Program, Cancer Research Centre (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) Instituto de salud Carlos III, Madrid, Spain
| | - Juan Torres Canizales
- Clinical Immunology Department, La Paz University Hospital and Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ) and Center for Biomedical Network Research on Rare Diseases (CIBERER U767), Madrid, Spain
| | - Martín Pérez-Andrés
- Clinical and Translation Research Program, Cancer Research Centre (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) Instituto de salud Carlos III, Madrid, Spain
| | - Elena Blanco
- Clinical and Translation Research Program, Cancer Research Centre (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) Instituto de salud Carlos III, Madrid, Spain
| | - Marjolein Wentink
- Department of Immunology, Erasmus University Medical Center (Erasmus MC), Rotterdam, Netherlands
| | - Carolien Bonroy
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Jana Nechvatalova
- Department of Allergology and Clinical Immunology, Faculty of Medicine, Masaryk University and St Anne's University Hospital in Brno, Brno, Czechia
| | - Tomas Milota
- Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Anne-Kathrin Kienzler
- Nuffield Department of Medicine, Experimental Medicine Division, University of Oxford, Oxford, United Kingdom
| | - Jan Philippé
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Ana E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mirjam van der Burg
- Department of Pediatrics, Laboratory for Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Tomas Kalina
- CLIP - Childhood Leukemia Investigation Prague, Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Alberto Orfao
- Clinical and Translation Research Program, Cancer Research Centre (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) Instituto de salud Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Lhermitte L, Barreau S, Morf D, Fernandez P, Grigore G, Barrena S, de Bie M, Flores-Montero J, Brüggemann M, Mejstrikova E, Nierkens S, Burgos L, Caetano J, Gaipa G, Buracchi C, da Costa ES, Sedek L, Szczepański T, Aanei CM, van der Sluijs-Gelling A, Delgado AH, Fluxa R, Lecrevisse Q, Pedreira CE, van Dongen JJM, Orfao A, van der Velden VHJ. Automated identification of leukocyte subsets improves standardization of database-guided expert-supervised diagnostic orientation in acute leukemia: a EuroFlow study. Mod Pathol 2021; 34:59-69. [PMID: 32999413 PMCID: PMC7806506 DOI: 10.1038/s41379-020-00677-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
Precise classification of acute leukemia (AL) is crucial for adequate treatment. EuroFlow has previously designed an AL orientation tube (ALOT) to guide toward the relevant classification panel and final diagnosis. In this study, we designed and validated an algorithm for automated (database-supported) gating and identification (AGI tool) of cell subsets within samples stained with ALOT. A reference database of normal peripheral blood (PB, n = 41) and bone marrow (BM; n = 45) samples analyzed with the ALOT was constructed, and served as a reference for the AGI tool to automatically identify normal cells. Populations not unequivocally identified as normal cells were labeled as checks and were classified by an expert. Additional normal BM (n = 25) and PB (n = 43) and leukemic samples (n = 109), analyzed in parallel by experts and the AGI tool, were used to evaluate the AGI tool. Analysis of normal PB and BM samples showed low percentages of checks (<3% in PB, <10% in BM), with variations between different laboratories. Manual analysis and AGI analysis of normal and leukemic samples showed high levels of correlation between cell numbers (r2 > 0.95 for all cell types in PB and r2 > 0.75 in BM) and resulted in highly concordant classification of leukemic cells by our previously published automated database-guided expert-supervised orientation tool for immunophenotypic diagnosis and classification of acute leukemia (Compass tool). Similar data were obtained using alternative, commercially available tubes, confirming the robustness of the developed tools. The AGI tool represents an innovative step in minimizing human intervention and requirements in expertise, toward a "sample-in and result-out" approach which may result in more objective and reproducible data analysis and diagnostics. The AGI tool may improve quality of immunophenotyping in individual laboratories, since high percentages of checks in normal samples are an alert on the quality of the internal procedures.
Collapse
Affiliation(s)
- Ludovic Lhermitte
- grid.508487.60000 0004 7885 7602Institut Necker-Enfants Malades, Institut National de Recherche Médicale U1151, Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Université de Paris, Paris, France
| | - Sylvain Barreau
- grid.508487.60000 0004 7885 7602Institut Necker-Enfants Malades, Institut National de Recherche Médicale U1151, Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Université de Paris, Paris, France
| | - Daniela Morf
- grid.413357.70000 0000 8704 3732FACS/Stem Cell Laboratory, Kantonsspital Aarau, Aarau, Switzerland
| | - Paula Fernandez
- grid.413357.70000 0000 8704 3732FACS/Stem Cell Laboratory, Kantonsspital Aarau, Aarau, Switzerland
| | | | - Susana Barrena
- Cytognos SL, Salamanca, Spain ,Translational and Clinical Research Program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain ,grid.11762.330000 0001 2180 1817Department of Medicine, University of Salamanca (USAL), Salamanca, Spain ,grid.452531.4Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain ,grid.413448.e0000 0000 9314 1427Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Maaike de Bie
- grid.5645.2000000040459992XDepartment of Immunology, Laboratory for Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain ,grid.11762.330000 0001 2180 1817Department of Medicine, University of Salamanca (USAL), Salamanca, Spain ,grid.452531.4Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain ,grid.413448.e0000 0000 9314 1427Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Monika Brüggemann
- grid.412468.d0000 0004 0646 2097Department of Hematology, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ester Mejstrikova
- grid.4491.80000 0004 1937 116XDepartment of Pediatric Hematology and Oncology, University Hospital Motol, Charles University, Prague, Czechia
| | - Stefan Nierkens
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Leire Burgos
- grid.411730.00000 0001 2191 685XApplied Medical Research Center (CIMA), IDISNA, Clinica Universidad de Navarra (UNAV), Pamplona, Spain
| | - Joana Caetano
- grid.418711.a0000 0004 0631 0608Hemato-Oncology Laboratory, Portuguese Institute of Oncology, Lisbon, Portugal
| | - Giuseppe Gaipa
- grid.7563.70000 0001 2174 1754Tettamanti Research Center, Pediatric Clinic University of Milano Bicocca, Monza, MB Italy
| | - Chiara Buracchi
- grid.7563.70000 0001 2174 1754Tettamanti Research Center, Pediatric Clinic University of Milano Bicocca, Monza, MB Italy
| | - Elaine Sobral da Costa
- grid.8536.80000 0001 2294 473XPediatrics Institute IPPMG, Faculty of Medicine, Federal University of Rio de Janeiro, Av. Horacio Macedo, Predio do CT, CEP, Rio de Janeiro, 21941-914 Brazil
| | - Lukasz Sedek
- grid.411728.90000 0001 2198 0923Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Tomasz Szczepański
- grid.411728.90000 0001 2198 0923Department of Pediatric Hematology and Oncology, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Carmen-Mariana Aanei
- grid.412954.f0000 0004 1765 1491Laboratory of Hematology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Alita van der Sluijs-Gelling
- grid.10419.3d0000000089452978Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Alejandro Hernández Delgado
- Cytognos SL, Salamanca, Spain ,Translational and Clinical Research Program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain ,grid.11762.330000 0001 2180 1817Department of Medicine, University of Salamanca (USAL), Salamanca, Spain ,grid.452531.4Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain ,grid.413448.e0000 0000 9314 1427Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Quentin Lecrevisse
- Translational and Clinical Research Program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain ,grid.11762.330000 0001 2180 1817Department of Medicine, University of Salamanca (USAL), Salamanca, Spain ,grid.452531.4Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain ,grid.413448.e0000 0000 9314 1427Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos E. Pedreira
- grid.8536.80000 0001 2294 473XSystems and Computing Department (PESC), COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Jacques J. M. van Dongen
- grid.10419.3d0000000089452978Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Centre (IBMCC, CSIC-USAL), Cytometry Service, NUCLEUS, Salamanca, Spain ,grid.11762.330000 0001 2180 1817Department of Medicine, University of Salamanca (USAL), Salamanca, Spain ,grid.452531.4Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain ,grid.413448.e0000 0000 9314 1427Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Vincent H. J. van der Velden
- grid.5645.2000000040459992XDepartment of Immunology, Laboratory for Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | |
Collapse
|
23
|
Linskens E, Diks AM, Neirinck J, Perez-Andres M, De Maertelaere E, Berkowska MA, Kerre T, Hofmans M, Orfao A, van Dongen JJM, Haerynck F, Philippé J, Bonroy C. Improved Standardization of Flow Cytometry Diagnostic Screening of Primary Immunodeficiency by Software-Based Automated Gating. Front Immunol 2020; 11:584646. [PMID: 33224147 PMCID: PMC7667243 DOI: 10.3389/fimmu.2020.584646] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/12/2020] [Indexed: 01/08/2023] Open
Abstract
Background Multiparameter flow cytometry (FC) is essential in the diagnostic work-up and classification of primary immunodeficiency (PIDs). The EuroFlow PID Orientation tube (PIDOT) allows identification of all main lymphocyte subpopulations in blood. To standardize data analysis, tools for Automated Gating and Identification (AG&I) of the informative cell populations, were developed by EuroFlow. Here, we evaluated the contribution of these innovative AG&I tools to the standardization of FC in the diagnostic work-up of PID, by comparing AG&I against expert-based (EuroFlow-standardized) Manual Gating (MG) strategy, and its impact on the reproducibility and clinical interpretation of results. Methods FC data files from 44 patients (13 CVID, 12 PID, 19 non-PID) and 26 healthy donor (HD) blood samples stained with PIDOT were analyzed in parallel by MG and AG&I, using Infinicyt™ software (Cytognos). For comparison, percentage differences in absolute cell counts/µL were calculated for each lymphocyte subpopulation. Data files showing differences >20% were checked for their potential clinical relevance, based on age-matched percentile (p5-p95) reference ranges. In parallel, intra- and inter-observer reproducibility of MG vs AG&I were evaluated in a subset of 12 samples. Results The AG&I approach was able to identify the vast majority of lymphoid events (>99%), associated with a significantly higher intra- and inter-observer reproducibility compared to MG. For most HD (83%) and patient (68%) samples, a high degree of agreement (<20% numerical differences in absolute cell counts/µL) was obtained between MG and the AG&I module. This translated into a minimal impact (<5% of observations) on the final clinical interpretation. In all except three samples, extended expert revision of the AG&I approach revealed no error. In the three remaining samples aberrant maturation and/or abnormal marker expression profiles were seen leading in all three cases to numerical alarms by AG&I. Conclusion Altogether, our results indicate that replacement of MG by the AG&I module would be associated with a greater reproducibility and robustness of results in the diagnostic work-up of patients suspected of PID. However, expert revision of the results of AG&I of PIDOT data still remains necessary in samples with numerical alterations and aberrant B- and T-cell maturation and/or marker expression profiles.
Collapse
Affiliation(s)
- Eleni Linskens
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Annieck M Diks
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Jana Neirinck
- Department of Diagnostic Science, Ghent University, Ghent, Belgium
| | - Martín Perez-Andres
- Cancer Research Centre (IBMCC, USAL-CSIC; CIBERONC CB16/12/00400), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca (USAL), Salamanca, Spain.,Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
| | | | - Magdalena A Berkowska
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Tessa Kerre
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Mattias Hofmans
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Alberto Orfao
- Cancer Research Centre (IBMCC, USAL-CSIC; CIBERONC CB16/12/00400), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca (USAL), Salamanca, Spain.,Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
| | - Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Filomeen Haerynck
- Department of Pediatric Pulmonology and Immunology and PID Research Laboratory, Ghent University Hospital, Ghent, Belgium
| | - Jan Philippé
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Department of Diagnostic Science, Ghent University, Ghent, Belgium
| | - Carolien Bonroy
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Department of Diagnostic Science, Ghent University, Ghent, Belgium
| |
Collapse
|
24
|
Henriques-Pons A, Beatrici CP, Sánchez-Arcila JC, da Silva FAB. Multiparametric Color Tendency Analysis (MCTA): A Method to Analyze Several Flow Cytometry Labelings Simultaneously. Front Bioeng Biotechnol 2020; 8:526814. [PMID: 33042962 PMCID: PMC7527824 DOI: 10.3389/fbioe.2020.526814] [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: 02/25/2020] [Accepted: 08/26/2020] [Indexed: 11/13/2022] Open
Abstract
Despite the remarkable evolution of flow cytometers, fluorescent probes, and flow cytometry analysis software, most users still follow the same ways for data analysis. Conventional flow cytometry analysis relies on the creation of dot plot sequences, based on two fluorescence parameters at a time, to evidence phenotypically distinct populations. Thus, reaching conclusions about the biological characteristics of the samples is a fragmented and challenging process. We present here the MCTA (Multiparametric Color Tendency Analysis), a method for data analysis that considers multiple labelings simultaneously, extending and complementing conventional analysis. The MCTA method executes the background fluorescence exclusion, spillover compensation, and a user-defined gating strategy for subpopulation analysis. The results are then presented in conventional FSC x SSC dot plots with statistical data. For each event, the method converts each of the multiple fluorescence colors under analysis into a vector, with longer vectors being attributed to more intense labelings. Then, the MCTA generates a resultant vector, which is therefore mostly influenced by predominant labelings. The radial position of this resultant vector corresponds to a resultant color, making it easy to visualize phenotypic modulations among cellular subpopulations. Besides, it is a deterministic method that quickly assigns a resulting color to all events that obey the gating strategy, with no polymeric regions defined by the user or downsampling. The MCTA application generates a single dot plot showing all events in the FCS file, but a resultant color is attributed only to those that obey the gating strategy. Therefore, it can also help to evidence rare events or unpredicted subpopulations naturally excluded from the regions defined by the user. We believe that the MCTA method adds a new perspective over multiparametric flow cytometry analysis while evidencing modulations of molecular labeling profiles based on multiple fluorescences. Availability and implementation: The instructions for the MCTA application is freely available at https://github.com/flowcytometry/MCTA.
Collapse
Affiliation(s)
- Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- *Correspondence: Andrea Henriques-Pons,
| | - Carine P. Beatrici
- Scientific Computing Program, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
25
|
Jacqmin H, Chatelain B, Louveaux Q, Jacqmin P, Dogné JM, Graux C, Mullier F. Clustering and Kernel Density Estimation for Assessment of Measurable Residual Disease by Flow Cytometry. Diagnostics (Basel) 2020; 10:E317. [PMID: 32443428 PMCID: PMC7277972 DOI: 10.3390/diagnostics10050317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/04/2022] Open
Abstract
Standardization, data mining techniques, and comparison to normality are changing the landscape of multiparameter flow cytometry in clinical hematology. On the basis of these principles, a strategy was developed for measurable residual disease (MRD) assessment. Herein, suspicious cell clusters are first identified at diagnosis using a clustering algorithm. Subsequently, automated multidimensional spaces, named "Clouds", are created around these clusters on the basis of density calculations. This step identifies the immunophenotypic pattern of the suspicious cell clusters. Thereafter, using reference samples, the "Abnormality Ratio" (AR) of each Cloud is calculated, and major malignant Clouds are retained, known as "Leukemic Clouds" (L-Clouds). In follow-up samples, MRD is identified when more cells fall into a patient's L-Cloud compared to reference samples (AR concept). This workflow was applied on simulated data and real-life leukemia flow cytometry data. On simulated data, strong patient-dependent positive correlation (R2 = 1) was observed between the AR and spiked-in leukemia cells. On real patient data, AR kinetics was in line with the clinical evolution for five out of six patients. In conclusion, we present a convenient flow cytometry data analysis approach for the follow-up of hematological malignancies. Further evaluation and validation on more patient samples and different flow cytometry panels is required before implementation in clinical practice.
Collapse
Affiliation(s)
- Hugues Jacqmin
- Hematology Laboratory, NAmur Research Institute for LIfe Sciences (NARILIS), Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université catholique de Louvain, 5530 Yvoir, Belgium; (B.C.); (F.M.)
| | - Bernard Chatelain
- Hematology Laboratory, NAmur Research Institute for LIfe Sciences (NARILIS), Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université catholique de Louvain, 5530 Yvoir, Belgium; (B.C.); (F.M.)
| | | | | | | | - Carlos Graux
- Department of Hematology, Namur Research Institute for Life Sciences (NARILIS), Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université catholique de Louvain, 5530 Yvoir, Belgium;
| | - François Mullier
- Hematology Laboratory, NAmur Research Institute for LIfe Sciences (NARILIS), Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université catholique de Louvain, 5530 Yvoir, Belgium; (B.C.); (F.M.)
| |
Collapse
|
26
|
Botafogo V, Pérez-Andres M, Jara-Acevedo M, Bárcena P, Grigore G, Hernández-Delgado A, Damasceno D, Comans S, Blanco E, Romero A, Arriba-Méndez S, Gastaca-Abasolo I, Pedreira CE, van Gaans-van den Brink JAM, Corbiere V, Mascart F, van Els CACM, Barkoff AM, Mayado A, van Dongen JJM, Almeida J, Orfao A. Age Distribution of Multiple Functionally Relevant Subsets of CD4+ T Cells in Human Blood Using a Standardized and Validated 14-Color EuroFlow Immune Monitoring Tube. Front Immunol 2020; 11:166. [PMID: 32174910 PMCID: PMC7056740 DOI: 10.3389/fimmu.2020.00166] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
CD4+ T cells comprise multiple functionally distinct cell populations that play a key role in immunity. Despite blood monitoring of CD4+ T-cell subsets is of potential clinical utility, no standardized and validated approaches have been proposed so far. The aim of this study was to design and validate a single 14-color antibody combination for sensitive and reproducible flow cytometry monitoring of CD4+ T-cell populations in human blood to establish normal age-related reference values and evaluate the presence of potentially altered profiles in three distinct disease models-monoclonal B-cell lymphocytosis (MBL), systemic mastocytosis (SM), and common variable immunodeficiency (CVID). Overall, 145 blood samples from healthy donors were used to design and validate a 14-color antibody combination based on extensive reagent testing in multiple cycles of design-testing-evaluation-redesign, combined with in vitro functional studies, gene expression profiling, and multicentric evaluation of manual vs. automated gating. Fifteen cord blood and 98 blood samples from healthy donors (aged 0-89 years) were used to establish reference values, and another 25 blood samples were evaluated for detecting potentially altered CD4 T-cell subset profiles in MBL (n = 8), SM (n = 7), and CVID (n = 10). The 14-color tube can identify ≥89 different CD4+ T-cell populations in blood, as validated with high multicenter reproducibility, particularly when software-guided automated (vs. manual expert-based) gating was used. Furthermore, age-related reference values were established, which reflect different kinetics for distinct subsets: progressive increase of naïve T cells, T-helper (Th)1, Th17, follicular helper T (TFH) cells, and regulatory T cells (Tregs) from birth until 2 years, followed by a decrease of naïve T cells, Th2, and Tregs in older children and a subsequent increase in multiple Th-cell subsets toward late adulthood. Altered and unique CD4+ T-cell subset profiles were detected in two of the three disease models evaluated (SM and CVID). In summary, the EuroFlow immune monitoring TCD4 tube allows fast, automated, and reproducible identification of ≥89 subsets of CD4+ blood T cells, with different kinetics throughout life. These results set the basis for in-depth T-cell monitoring in different disease and therapeutic conditions.
Collapse
Affiliation(s)
- Vitor Botafogo
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martín Pérez-Andres
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - María Jara-Acevedo
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Sequencing Service, NUCLEUS, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Paloma Bárcena
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Alejandro Hernández-Delgado
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Cytognos SL, Salamanca, Spain
| | - Daniela Damasceno
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Suzanne Comans
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Elena Blanco
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Romero
- Miguel Armijo Primary Health Care Centre, Sanidad de Castilla y León (SACYL), Salamanca, Spain
| | | | - Irene Gastaca-Abasolo
- Gynecology and Obstetrics Service, University Hospital of Salamanca, Salamanca, Spain
| | - Carlos Eduardo Pedreira
- Systems and Computing Department (PESC), COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Véronique Corbiere
- Laboratory of Vaccinology and Mucosal Immunity, Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Françoise Mascart
- Laboratory of Vaccinology and Mucosal Immunity, Université libre de Bruxelles (ULB), Brussels, Belgium
- Immunobiology Clinic, Hôpital Erasme, Brussels, Belgium
| | - Cécile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Alex-Mikael Barkoff
- Institute of Biomedicine, Department of Microbiology, Virology and Immunology, University of Turku (UTU), Turku, Finland
| | - Andrea Mayado
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Jacques J. M. van Dongen
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Julia Almeida
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
27
|
Zelm MC, McKenzie CI, Varese N, Rolland JM, O'Hehir RE. Recent developments and highlights in immune monitoring of allergen immunotherapy. Allergy 2019; 74:2342-2354. [PMID: 31587309 DOI: 10.1111/all.14078] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
Abstract
Allergic diseases are the most common chronic immune-mediated disorders and can manifest with an enormous diversity in clinical severity and symptoms. Underlying mechanisms for the adverse immune response to allergens and its downregulation by treatment are still being revealed. As a result, there have been, and still are, major challenges in diagnosis, prediction of disease progression/evolution and treatment. Currently, the only corrective treatment available is allergen immunotherapy (AIT). AIT modifies the immune response through long-term repeated exposure to defined doses of allergen. However, as the treatment usually needs to be continued for several years to be effective, and can be accompanied by adverse reactions, many patients face difficulties completing their schedule. Long-term therapy also potentially incurs high costs. Therefore, there is a great need for objective markers to predict or to monitor individual patient's beneficial changes in immune response during therapy so that efficacy can be identified as early as possible. In this review, we specifically address recent technical developments that have generated new insights into allergic disease pathogenesis, and how these could potentially be translated into routine laboratory assays for disease monitoring during AIT that are relatively inexpensive, robust and scalable.
Collapse
Affiliation(s)
- Menno C. Zelm
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Craig I. McKenzie
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
| | - Nirupama Varese
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Jennifer M. Rolland
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Robyn E. O'Hehir
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| |
Collapse
|
28
|
van Dongen JJM, O'Gorman MRG, Orfao A. EuroFlow and its activities: Introduction to the special EuroFlow issue of The Journal of Immunological Methods. J Immunol Methods 2019; 475:112704. [PMID: 31758969 DOI: 10.1016/j.jim.2019.112704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, the Netherlands.
| | - Maurice R G O'Gorman
- Departments of Pathology and Pediatrics, The Keck School of Medicine, U. of Southern California, Children's Hospital of Los Angeles Los Angeles, CA, USA
| | - Alberto Orfao
- Cancer Research Centre (IBMCC-CASIC/USAL), Department of Medicine, Cytometry Service (NUCLEUS) and Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca (Spain) and CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Maurillo L, Bassan R, Cascavilla N, Ciceri F. Quality of Response in Acute Myeloid Leukemia: The Role of Minimal Residual Disease. Cancers (Basel) 2019; 11:cancers11101417. [PMID: 31548502 PMCID: PMC6826465 DOI: 10.3390/cancers11101417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022] Open
Abstract
In the acute myeloid leukemia (AML) setting, research has extensively investigated the existence and relevance of molecular biomarkers, in order to better tailor therapy with newly developed agents and hence improve outcomes and/or save the patient from poorly effective therapies. In particular, in patients with AML, residual disease after therapy does reflect the sum of the contributions of all factors associated with diagnosis and post-diagnosis resistance. The evaluation of minimal/measurable residual disease (MRD) can be considered as a key tool to guide patient’s management and a promising endpoint for clinical trials. In this narrative review, we discuss MRD evaluation as biomarker for tailored therapy in AML patients; we briefly report current evidence on the use of MRD in clinical practice, and comment on the potential ability of MRD in the assessment of the efficacy of new molecules.
Collapse
Affiliation(s)
- Luca Maurillo
- Hematology Unit, Department of Biomedicine and Prevention, Fondazione Policlinico Tor Vergata, Hospital, 00133 Rome, Italy.
| | - Renato Bassan
- Hematology Unit, dell'Angelo Hospital and Santissimi Giovanni and Paolo Hospital, 30174 Mestre and Venice, Italy.
| | - Nicola Cascavilla
- Hematology Unit, Onco-hematology Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy.
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, IRCCS S. Raffaele Scientific Institution, 20132 Milan, Italy.
| |
Collapse
|
30
|
EuroFlow Lymphoid Screening Tube (LST) data base for automated identification of blood lymphocyte subsets. J Immunol Methods 2019; 475:112662. [PMID: 31454495 DOI: 10.1016/j.jim.2019.112662] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/31/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023]
Abstract
In recent years the volume and complexity of flow cytometry data has increased substantially. This has led to a greater number of identifiable cell populations in a single measurement. Consequently, new gating strategies and new approaches for cell population definition are required. Here we describe how the EuroFlow Lymphoid Screening Tube (LST) reference data base for peripheral blood (PB) samples was designed, constructed and validated for automated gating of the distinct lymphoid (and myeloid) subsets in PB of patients with chronic lymphoproliferative disorders (CLPD). A total of 46 healthy/reactive PB samples which fulfilled pre-defined technical requirements, were used to construct the LST-PB reference data base. In addition, another set of 92 PB samples (corresponding to 10 healthy subjects, 51 B-cell CLPD and 31 T/NK-cell CLPD patients), were used to validate the automated gating and cell-population labeling tools with the Infinicyt software. An overall high performance of the LST-PB data base was observed with a median percentage of alarmed cellular events of 0.8% in 10 healthy donor samples and of 44.4% in CLPD data files containing 49.8% (range: 1.3-96%) tumor cells. The higher percent of alarmed cellular events in every CLPD sample was due to aberrant phenotypes (75.6% cases) and/or to abnormally increased cell counts (86.6% samples). All 18 (22%) data files that only displayed numerical alterations, corresponded to T/NK-cell CLPD cases which showed a lower incidence of aberrant phenotypes (41%) vs B-cell CLPD cases (100%). Comparison between automated vs expert-bases manual classification of normal (r2 = 0.96) and tumor cell populations (rho = 0.99) showed a high degree of correlation. In summary, our results show that automated gating of cell populations based on the EuroFlow LST-PB data base provides an innovative, reliable and reproducible tool for fast and simplified identification of normal vs pathological B and T/NK lymphocytes in PB of CLPD patients.
Collapse
|