1
|
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
|
2
|
Cools M, Grijp C, Neirinck J, Tavernier SJ, Schelstraete P, Van De Velde J, Morbée L, De Baere E, Bonroy C, van Bever Y, Bruggenwirth H, Vermont C, Hannema SE, De Rijke Y, Abdulhadi-Atwan M, Zangen D, Verdin H, Haerynck F. Spleen function is reduced in individuals with NR5A1 variants with or without a difference of sex development: a cross-sectional study. Eur J Endocrinol 2024; 190:34-43. [PMID: 38128121 DOI: 10.1093/ejendo/lvad174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE NR5A1 is a key regulator of sex differentiation and has been implicated in spleen development through transcription activation of TLX1. Concerns exist about hypo- or asplenism in individuals who have a difference of sex development (DSD) due to an NR5A1 disease-causing variant. We aimed to assess spleen anatomy and function in a clinical cohort of such individuals and in their asymptomatic family member carriers. DESIGN Cross-sectional assessment in 22 patients with a DSD or primary ovarian insufficiency and 5 asymptomatic carriers from 18 families, harboring 14 different NR5A1 variants. METHODS Spleen anatomy was assessed by ultrasound, spleen function by peripheral blood cell count, white blood cell differentiation, percentage of nonswitched memory B cells, specific pneumococcal antibody response, % pitted red blood cells, and Howell-Jolly bodies. RESULTS Patients and asymptomatic heterozygous individuals had significantly decreased nonswitched memory B cells compared to healthy controls, but higher than asplenic patients. Thrombocytosis and spleen hypoplasia were present in 50% of heterozygous individuals. Four out of 5 individuals homozygous for the previously described p.(Arg103Gln) variant had asplenia. CONCLUSIONS Individuals harboring a heterozygous NR5A1 variant that may cause DSD have a considerable risk for functional hyposplenism, irrespective of their gonadal phenotype. Splenic function should be assessed in these individuals, and if affected or unknown, prophylaxis is recommended to prevent invasive encapsulated bacterial infections. The splenic phenotype associated with NR5A1 variants is more severe in homozygous individuals and is, at least for the p.(Arg103Gln) variant, associated with asplenism.
Collapse
Affiliation(s)
- Martine Cools
- Department of Internal Medicine and Pediatrics, Pediatric Endocrinology Service, Ghent University, Ghent University Hospital, 9000 Ghent, Belgium
| | - Celien Grijp
- Department of Internal Medicine and Pediatrics, Pediatric Endocrinology Service, Ghent University, Ghent University Hospital, 9000 Ghent, Belgium
| | - Jana Neirinck
- Department of Diagnostic Science, Ghent University, Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Simon J Tavernier
- Department of Internal Medicine and Pediatrics, PID Research Lab, Ghent University, 9000 Ghent, Belgium
- Laboratory of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Petra Schelstraete
- Department of Internal Medicine and Pediatrics, Pediatric Pulmonology and Infectious Diseases, Ghent University, Ghent University Hospital, 9000 Ghent, Belgium
| | - Julie Van De Velde
- Department of Internal Medicine and Pediatrics, Pediatric Endocrinology Service, Ghent University, Ghent University Hospital, 9000 Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lieve Morbée
- Department of Radiology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University Hospital, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Science, Ghent University, Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Yolande van Bever
- Department of Clinical Genetics, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Hennie Bruggenwirth
- Department of Clinical Genetics, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Clementien Vermont
- Department of Pediatric Infectious Diseases and Immunology, Erasmus Medical Center-Sophia Children's Hospital, 3015 GD Rotterdam, The Netherlands
| | - Sabine E Hannema
- Department of Pediatric Endocrinology, Erasmus Medical Center-Sophia Children's Hospital, 3015 GD Rotterdam, The Netherlands
- Department of Paediatric Endocrinology, Gastroenterology Endocrinology Metabolism, Reproduction and Development, Amsterdam UMC location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolanda De Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center 3015 GD Rotterdam, The Netherlands
| | - Maha Abdulhadi-Atwan
- Department of Pediatrics, Pediatric Endocrinology Service, Palestine Red Crescent Society Hospital, PO Box 421, Hebron, Palestine
| | - David Zangen
- Division of Pediatric Endocrinology, Faculty of Medicine, Hadassah University Hospital, Hebrew University of Jerusalem, 91120 Jerusalem, Israel
| | - Hannah Verdin
- Center for Medical Genetics, Ghent University Hospital, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, PID Research Lab, Ghent University, 9000 Ghent, Belgium
- Department of Pediatric Pulmonology and Immunology, Centre for Primary Immune Deficiency, Jeffrey Modell Diagnostic and Research Centre for PID, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
3
|
Pérez-Escurza O, Flores-Montero J, Óskarsson JÞ, Sanoja-Flores L, Del Pozo J, Lecrevisse Q, Martín S, Reed ER, Hákonardóttir GK, Harding S, Þorsteinsdóttir S, Rögnvaldsson S, Love TJ, Durie B, Kristinsson SY, Orfao A. Immunophenotypic assessment of clonal plasma cells and B-cells in bone marrow and blood in the diagnostic classification of early stage monoclonal gammopathies: an iSTOPMM study. Blood Cancer J 2023; 13:182. [PMID: 38072838 PMCID: PMC10711003 DOI: 10.1038/s41408-023-00944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Monoclonal gammopathy of undetermined significance (MGUS) is the earliest discernible stage of multiple myeloma (MM) and Waldenström's macroglobulinemia (WM). Early diagnosis of MG may be compromised by the low-level infiltration, undetectable to low-sensitive methodologies. Here, we investigated the prevalence and immunophenotypic profile of clonal (c) plasma cells (PC) and/or cB-lymphocytes in bone marrow (BM) and blood of subjects with a serum M-component from the iSTOPMM program, using high-sensitive next-generation flow cytometry (NGF), and its utility in the diagnostic classification of early-stage MG. We studied 164 paired BM and blood samples from 82 subjects, focusing the analysis on: 55 MGUS, 12 smoldering MM (SMM) and 8 smoldering WM (SWM). cPC were detected in 84% of the BM samples and cB-lymphocytes in 45%, coexisting in 39% of cases. In 29% of patients, the phenotypic features of cPC and/or cB-lymphocytes allowed a more accurate disease classification, including: 19/55 (35%) MGUS, 1/12 (8%) SMM and 2/8 (25%) SWM. Blood samples were informative in 49% of the BM-positive cases. We demonstrated the utility of NGF for a more accurate diagnostic classification of early-stage MG.
Collapse
Affiliation(s)
- Oihane Pérez-Escurza
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, 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
| | | | - Luzalba Sanoja-Flores
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Biomedicine of Seville, Department of Hematology, University Hospital Virgen del Rocío of the Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Seville, Spain
| | - Julio Del Pozo
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Quentin Lecrevisse
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, 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
| | - Silvia Martín
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Elín Ruth Reed
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | | | - Sigrún Þorsteinsdóttir
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Rigshospitalet, Copenhagen, Denmark
| | - Sæmundur Rögnvaldsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Thorvardur Jon Love
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Brian Durie
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sigurður Yngvi Kristinsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, 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
|
4
|
Criado I, Nieto WG, Oliva-Ariza G, Fuentes-Herrero B, Teodosio C, Lecrevisse Q, Lopez A, Romero A, Almeida J, Orfao A. Age- and Sex-Matched Normal Leukocyte Subset Ranges in the General Population Defined with the EuroFlow Lymphocyte Screening Tube (LST) for Monoclonal B-Cell Lymphocytosis (MBL) vs. Non-MBL Subjects. Cancers (Basel) 2022; 15:cancers15010058. [PMID: 36612056 PMCID: PMC9817826 DOI: 10.3390/cancers15010058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/02/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Reference ranges of blood-circulating leukocyte populations by, e.g., age and sex, are required for monitoring immune-cell kinetics. Most previous reports in which flow cytometry has been used to define the reference ranges for leukocyte counts included a limited number of donors and/or cell populations and/or did not consider age and sex simultaneously. Moreover, other factors not previously considered in the definition of normal ranges, such as the presence of chronic-lymphocytic-leukemia (CLL)-like low-count monoclonal B-cell lymphocytosis (MBLlo), might also be associated with an altered distribution of leukocytes in blood in association with an immunodeficiency and increased risk of infection and cancer. Here, we established reference cell-count ranges for the major populations of leukocytes in blood of non-MBL and MBLlo adult Caucasians matched by age and sex using the EuroFlow Lymphocyte Screening Tube (LST). A total of 706 Caucasian adult donors—622 non-MBL and 84 MBLlo—were recruited from the general population. Among non-MBL donors, the total leukocyte, neutrophil, basophil dendritic cell and monocyte counts remained stable through adulthood, while the absolute numbers of T- and B-cell populations and plasma cells decreased with age. The number of eosinophils and NK-cell increased over time, with clear differences according to sex for certain age ranges. In MBLlo subjects, few differences in the absolute cell counts by age (vs. non-MBL) were observed, and MBLlo men and women showed similar trends to non-MBL subjects except for the B-cell count drop observed in >70 y-men, which was more pronounced in MBLlo vs. non-MBL controls. Building robust age- and sex-matched reference ranges for the most relevant immune-cell populations in the blood of non-MBL donors is essential to appropriately identify an altered immune status in different clinical settings and highlight the altered immune-cell profiles of MBLlo subjects.
Collapse
Affiliation(s)
- Ignacio Criado
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Wendy G. Nieto
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Guillermo Oliva-Ariza
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Blanca Fuentes-Herrero
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Cristina Teodosio
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Quentin Lecrevisse
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Antonio Lopez
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Alfonso Romero
- Centro de Atención Primaria de Salud Miguel Armijo, Sanidad de Castilla y León (SACyL), 37007 Salamanca, Spain
| | - Julia Almeida
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (J.A.); (A.O.); Tel.: +34-923-29-4811 (J.A.)
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC–Universidad de Salamanca); Cytometry Service, NUCLEUS; Departamento de Medicina, Universidad de Salamanca (https://ror.org/02f40zc51) and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (J.A.); (A.O.); Tel.: +34-923-29-4811 (J.A.)
| | | |
Collapse
|
5
|
Gorrese M, Bertolini A, Fresolone L, Campana A, Pezzullo L, Guariglia R, Mettivier L, Manzo P, Cuffa B, D'Alto F, Serio B, Selleri C, Giudice V. Inter-intra instrument comparison and standardization of a 10-color immunophenotyping for B and T cell non-Hodgkin lymphoma diagnosis and monitoring. J Immunol Methods 2022; 511:113374. [PMID: 36243108 DOI: 10.1016/j.jim.2022.113374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022]
Abstract
Harmonization of flow cytometry protocols from instrument settings to antibody panel and reagents is highly encouraged for inter-laboratory data comparison in both research and clinical settings, especially for minimal residual disease monitoring evaluation in hematological diseases across centers. Here, we described inter-intra instrument comparison of two standardized 10-color staining dried tubes for B- and T-cell lymphoproliferative disorder diagnosis and monitoring on two different flow cytometers, a Beckman Coulter NaviosEx and a Beckman Coulter DxFlex. A total of 47 consecutive patients were enrolled, and 39 of them were evaluable for further studies. We show highly comparable results between the two cytometers for cell frequency and fluorescence intensity signals for both standardized 10-color staining dried tubes. For this latter, fluorescence of each antibody and subject was normalized on the mean value obtained from the entire study cohort thus reducing the effects of biological variability and allowing comparison between instruments with different detector sensitivity. In summary, dried tubes were confirmed as an optimal standardized diagnostic tool, especially when associated with EuroFlow standardized procedures by minimizing technical and biological variability. However, data analysis is still operator-dependent, and more efforts are needed to develop automated or semi-automated software for flow cytometry data analysis for diagnostic purposes.
Collapse
Affiliation(s)
- Marisa Gorrese
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Angela Bertolini
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Lucia Fresolone
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Annapaola Campana
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Luca Pezzullo
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Roberto Guariglia
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Laura Mettivier
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Paola Manzo
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Bianca Cuffa
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Francesca D'Alto
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Bianca Serio
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Carmine Selleri
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy; Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy.
| | - Valentina Giudice
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy; Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
| |
Collapse
|
6
|
Raskovalova T, Scheffen L, Jacob MC, Chevalier S, Tondeur S, Bulabois B, Meunier M, Szymanski G, Lefebvre C, Planta C, Dumestre-Perard C, Gonnet N, Garban F, Merle R, Park S, Labarère J. Flow cytometry lyophilised-reagent tube for quantifying peripheral blood neutrophil myeloperoxidase expression in myelodysplastic syndromes (MPO-MDS-Develop): protocol for a diagnostic accuracy study. BMJ Open 2022; 12:e065850. [PMID: 36207039 PMCID: PMC9557768 DOI: 10.1136/bmjopen-2022-065850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Suspicion of myelodysplastic syndromes (MDS) is the most common reason for bone marrow aspirate in elderly patients. Peripheral blood neutrophil myeloperoxidase expression quantified by flow cytometric analysis might rule out MDS for up to 35% of patients referred for suspected disease, without requiring bone marrow aspiration. Yet laboratory-developed liquid antibody cocktails have practical limitations, because of lack of standardisation and poor stability. This research project aims to estimate the level of agreement and comparative accuracy between a single-use flow cytometry tube of lyophilised reagents (BD Lyotube Stain 468) and its laboratory-developed liquid reagent counterpart in quantifying peripheral blood neutrophil myeloperoxidase expression, among adult patients referred for suspected MDS. METHODS AND ANALYSIS The MPO-MDS-Develop project is a cross-sectional diagnostic accuracy study of two index tests by comparison with a reference standard in consecutive unselected adult patients conducted at a single university hospital. Flow cytometry analysis of peripheral blood samples will be performed by independent operators blinded to the reference diagnosis, using either Lyotube Stain 468 or laboratory-developed liquid reagent cocktail. The reference diagnosis of MDS will be established by cytomorphological evaluation of bone marrow aspirate by two independent haematopathologists blinded to the index test results. Morphologic assessment will be complemented by bone marrow flow cytometric score, karyotype and targeted next-generation sequencing panel of 43 genes, where relevant. The target sample size is 103 patients. ETHICS AND DISSEMINATION An institutional review board (Comité de Protection des Personnes Sud Est III, Lyon, France) approved the protocol prior to study initiation (reference number: 2020-028-B). Participants will be recruited using an opt-out approach. Efforts will be made to release the primary results within 6 months of study completion. TRIAL REGISTRATION NUMBER NCT04399018.
Collapse
Affiliation(s)
- Tatiana Raskovalova
- Laboratoire d'Immunologie, Grenoble Alpes University Hospital, Grenoble, France
- Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Univ. Grenoble Alpes, Grenoble, France
| | - Laura Scheffen
- Laboratoire d'Immunologie, Grenoble Alpes University Hospital, Grenoble, France
| | - Marie-Christine Jacob
- Laboratoire d'Immunologie, Grenoble Alpes University Hospital, Grenoble, France
- Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Univ. Grenoble Alpes, Grenoble, France
| | - Simon Chevalier
- Laboratoire d'Hématologie Biologique, Grenoble Alpes University Hospital, Grenoble, France
| | - Sylvie Tondeur
- Laboratoire d'Hématologie Biologique, Grenoble Alpes University Hospital, Grenoble, France
| | - Bénédicte Bulabois
- Laboratoire d'Hématologie Biologique, Grenoble Alpes University Hospital, Grenoble, France
| | - Mathieu Meunier
- Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Univ. Grenoble Alpes, Grenoble, France
- Clinique Universitaire d'Hématologie, Grenoble Alpes University Hospital, Grenoble, France
| | - Gautier Szymanski
- Laboratoire d'Hématologie Biologique, Grenoble Alpes University Hospital, Grenoble, France
| | - Christine Lefebvre
- Laboratoire d'Hématologie Biologique, Grenoble Alpes University Hospital, Grenoble, France
| | - Charlotte Planta
- Laboratoire d'Immunologie, Grenoble Alpes University Hospital, Grenoble, France
| | | | - Nicolas Gonnet
- CIC 1406, INSERM, Grenoble Alpes University Hospital, Univ. Grenoble Alpes, Grenoble, France
| | - Frédéric Garban
- Clinique Universitaire d'Hématologie, Grenoble Alpes University Hospital, Grenoble, France
- TIMC, UMR 5525, CNRS, Univ. Grenoble Alpes, Grenoble, France
| | - Raymond Merle
- Département Universitaire des Patients, Univ. Grenoble Alpes, Grenoble, France
| | - Sophie Park
- Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Univ. Grenoble Alpes, Grenoble, France
- Clinique Universitaire d'Hématologie, Grenoble Alpes University Hospital, Grenoble, France
| | - José Labarère
- TIMC, UMR 5525, CNRS, Univ. Grenoble Alpes, Grenoble, France
- Clinical Epidemiology Unit, Grenoble Alpes University Hospital, Grenoble, France
| |
Collapse
|
7
|
Zlei M, Sidorov IA, Joosten SA, Heemskerk MHM, Myeni SK, Pothast CR, de Brouwer CS, Boomaars-van der Zanden AL, van Meijgaarden KE, Morales ST, Wessels E, Janse JJ, Goeman JJ, Cobbaert CM, Kroes ACM, Cannegieter SC, Roestenberg M, Visser LG, Kikkert M, Feltkamp MCW, Arbous SM, Staal FJT, Ottenhoff THM, van Dongen JJM, Roukens AHE, de Vries JJC. Immune Determinants of Viral Clearance in Hospitalised COVID-19 Patients: Reduced Circulating Naïve CD4+ T Cell Counts Correspond with Delayed Viral Clearance. Cells 2022; 11:2743. [PMID: 36078151 PMCID: PMC9455062 DOI: 10.3390/cells11172743] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Virus-specific cellular and humoral responses are major determinants for protection from critical illness after SARS-CoV-2 infection. However, the magnitude of the contribution of each of the components to viral clearance remains unclear. Here, we studied the timing of viral clearance in relation to 122 immune parameters in 102 hospitalised patients with moderate and severe COVID-19 in a longitudinal design. Delayed viral clearance was associated with more severe disease and was associated with higher levels of SARS-CoV-2-specific (neutralising) antibodies over time, increased numbers of neutrophils, monocytes, basophils, and a range of pro-inflammatory cyto-/chemokines illustrating ongoing, partially Th2 dominating, immune activation. In contrast, early viral clearance and less critical illness correlated with the peak of neutralising antibodies, higher levels of CD4 T cells, and in particular naïve CD4+ T cells, suggesting their role in early control of SARS-CoV-2 possibly by proving appropriate B cell help. Higher counts of naïve CD4+ T cells also correlated with lower levels of MIF, IL-9, and TNF-beta, suggesting an indirect role in averting prolonged virus-induced tissue damage. Collectively, our data show that naïve CD4+ T cell play a critical role in rapid viral T cell control, obviating aberrant antibody and cytokine profiles and disease deterioration. These data may help in guiding risk stratification for severe COVID-19.
Collapse
Affiliation(s)
- Mihaela Zlei
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Igor A. Sidorov
- Clinical Microbiological Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Mirjam H. M. Heemskerk
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Sebenzile K. Myeni
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Cilia R. Pothast
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Caroline S. de Brouwer
- Clinical Microbiological Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - A. Linda Boomaars-van der Zanden
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Krista E. van Meijgaarden
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Shessy T. Morales
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Els Wessels
- Clinical Microbiological Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jacqueline J. Janse
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jelle J. Goeman
- Medical Statistics Section, Department of Biomedical Data Sciences, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Christa M. Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Aloys C. M. Kroes
- Clinical Microbiological Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Suzanne C. Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Leonardus G. Visser
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marjolein Kikkert
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Mariet C. W. Feltkamp
- Clinical Microbiological Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Sesmu M. Arbous
- Department of Clinical Epidemiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Intensive Care, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Frank J. T. Staal
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Anna H. E. Roukens
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jutte J. C. de Vries
- Clinical Microbiological Laboratory, Department of Medical Microbiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | | |
Collapse
|
8
|
Glencross DK, Swart L, Pretorius M, Lawrie D. Evaluation of fixed-panel, multicolour ClearLLab 10C at an academic flow cytometry laboratory in Johannesburg, South Africa. Afr J Lab Med 2022; 11:1458. [PMID: 35937760 PMCID: PMC9350555 DOI: 10.4102/ajlm.v11i1.1458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Background Flow cytometric immunophenotyping is well established for the diagnosis of haematological neoplasms. New commercially available systems offer fixed, pre-aliquoted multi-parameter analysis to simplify sample preparation and standardise data analysis. Objective The Beckman Coulter (BC) ClearLLab™ 10C (4-tube) system was evaluated against an existing laboratory developed test (LDT). Methods Peripheral blood and bone marrow aspirates (n = 101), tested between August 2019 and November 2019 at an academic pathology laboratory in Johannesburg, South Africa, were analysed. Following daily instrument quality control, samples were prepared for LDT (using > 20 2–4-colour in-house panels and an extensive liquid monoclonal reagent repertoire) or ClearLLab 10C, and respectively analysed using in-house protocols on a Becton Dickinson FACSCalibur, or manufacturer-directed protocols on a BC Navios. Becton Dickinson Paint-a-Gate or BC Kaluza C software facilitated data interpretation. Diagnostic accuracy (concordance) was established by calculating sensitivity and specificity outcomes. Results Excellent agreement (clinical diagnostic concordance) with 100% specificity and sensitivity was established between LDT and ClearLLab 10C in 67 patients with a haematological neoplasm and 34 participants with no haematological disease. Similar acceptable diagnostic concordance (97%) was noted when comparing ClearLLab 10C to clinicopathological outcomes. Additionally, the ClearLLab 10C panels, analysed with Kaluza C software, enabled simultaneous discrimination of disease and concurrent background myeloid and lymphoid haematological populations, including assessing stages of maturation or sub-populations. Conclusion ClearLLab 10C panels provide excellent agreement to existing LDTs and may reliably be used for immunophenotyping of haematological neoplasms, simplifying and standardising sample preparation and data acquisition.
Collapse
Affiliation(s)
- Deborah K Glencross
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, Charlotte Maxeke Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| | - Leanne Swart
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, Charlotte Maxeke Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| | - Melanie Pretorius
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, Charlotte Maxeke Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| | - Denise Lawrie
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, Charlotte Maxeke Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
9
|
Kužílková D, Puñet-Ortiz J, Aui PM, Fernández J, Fišer K, Engel P, van Zelm MC, Kalina T. Standardization of Workflow and Flow Cytometry Panels for Quantitative Expression Profiling of Surface Antigens on Blood Leukocyte Subsets: An HCDM CDMaps Initiative. Front Immunol 2022; 13:827898. [PMID: 35222411 PMCID: PMC8874145 DOI: 10.3389/fimmu.2022.827898] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background The Human Cell Differentiation Molecules (HCDM) organizes Human Leukocyte Differentiation Antigen (HLDA) workshops to test and name clusters of antibodies that react with a specific antigen. These cluster of differentiation (CD) markers have provided the scientific community with validated antibody clones, consistent naming of targets and reproducible identification of leukocyte subsets. Still, quantitative CD marker expression profiles and benchmarking of reagents at the single-cell level are currently lacking. Objective To develop a flow cytometric procedure for quantitative expression profiling of surface antigens on blood leukocyte subsets that is standardized across multiple research laboratories. Methods A high content framework to evaluate the titration and reactivity of Phycoerythrin (PE)-conjugated monoclonal antibodies (mAbs) was created. Two flow cytometry panels were designed: an innate cell tube for granulocytes, dendritic cells, monocytes, NK cells and innate lymphoid cells (12-color) and an adaptive lymphocyte tube for naive and memory B and T cells, including TCRγδ+, regulatory-T and follicular helper T cells (11-color). The potential of these 2 panels was demonstrated via expression profiling of selected CD markers detected by PE-conjugated antibodies and evaluated using 561 nm excitation. Results Using automated data annotation and dried backbone reagents, we reached a robust workflow amenable to processing hundreds of measurements in each experiment in a 96-well plate format. The immunophenotyping panels enabled discrimination of 27 leukocyte subsets and quantitative detection of the expression of PE-conjugated CD markers of interest that could quantify protein expression above 400 units of antibody binding capacity. Expression profiling of 4 selected CD markers (CD11b, CD31, CD38, CD40) showed high reproducibility across centers, as well as the capacity to benchmark unique clones directed toward the same CD3 antigen. Conclusion We optimized a procedure for quantitative expression profiling of surface antigens on blood leukocyte subsets. The workflow, bioinformatics pipeline and optimized flow panels enable the following: 1) mapping the expression patterns of HLDA-approved mAb clones to CD markers; 2) benchmarking new antibody clones to established CD markers; 3) defining new clusters of differentiation in future HLDA workshops.
Collapse
Affiliation(s)
- Daniela Kužílková
- Childhood Leukaemia Investigation Prague (CLIP), Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic and University Hospital Motol, Prague, Czechia
| | - Joan Puñet-Ortiz
- Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain
| | - Pei M. Aui
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Javier Fernández
- Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain
| | - Karel Fišer
- Childhood Leukaemia Investigation Prague (CLIP), Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic and University Hospital Motol, Prague, Czechia
| | - Pablo Engel
- Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain
| | - Menno C. van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Tomáš Kalina
- Childhood Leukaemia Investigation Prague (CLIP), Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic and University Hospital Motol, Prague, Czechia
- *Correspondence: Tomáš Kalina,
| |
Collapse
|
10
|
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
|
11
|
Sędek Ł, Flores-Montero J, van der Sluijs A, Kulis J, te Marvelde J, Philippé J, Böttcher S, Bitter M, Caetano J, van der Velden VHJ, Sonneveld E, Buracchi C, Santos AH, Lima M, Szczepański T, van Dongen JJM, Orfao A. Impact of Pre-Analytical and Analytical Variables Associated with Sample Preparation on Flow Cytometric Stainings Obtained with EuroFlow Panels. Cancers (Basel) 2022; 14:cancers14030473. [PMID: 35158741 PMCID: PMC8833630 DOI: 10.3390/cancers14030473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Objective interpretation of flow cytometry may be hampered by a lack of standardized sample preparation procedures. The EuroFlow consortium conducted a series of experiments to determine the potential impact of different pre-analytical and analytical factors on the variability of results in terms of relative cell populations distribution and marker expression levels. The experiments were performed on healthy donors and patients with different hematological malignancies (e.g., acute leukemia, lymphoma, multiple myeloma, and myelodysplastic syndrome) to mimic real-world clinical settings. Overall, the results showed that sample storage conditions, anticoagulant use, and sample processing protocol might need to be tailored for sample and cell type(s), as well as to the specific markers evaluated. However, defining of well-balanced boundaries for storage time to 24 h, staining-acquisition delay to 3 h, and choosing a washing buffer of pH within the range of 7.2 to 7.8 would be a valid recommendation for most applications and circumstances described herein. Abstract Objective interpretation of FC results may still be hampered by limited technical standardization. The EuroFlow consortium conducted a series of experiments to determine the impact of different variables on the relative distribution and the median fluorescence intensity (MFI) of markers stained on different cell populations, from both healthy donors and patients’ samples with distinct hematological malignancies. The use of different anticoagulants; the time interval between sample collection, preparation, and acquisition; pH of washing buffers; and the use of cell surface membrane-only (SM) vs. cell surface plus intracytoplasmic (SM+CY) staining protocols, were evaluated. Our results showed that only monocytes were represented at higher percentages in EDTA- vs. heparin-anticoagulated samples. Application of SM or SM+CY protocols resulted in slight differences in the percentage of neutrophils and debris determined only with particular antibody combinations. In turn, storage of samples for 24 h at RT was associated with greater percentage of debris and cell doublets when the plasma cell disorder panel was used. Furthermore, 24 h storage of stained cells at RT was selectively detrimental for MFI levels of CD19 and CD45 on mature B- and T-cells (but not on leukemic blasts, clonal B- and plasma cells, neutrophils, and NK cells). The obtained results showed that the variables evaluated might need to be tailored for sample and cell type(s) as well as to the specific markers compared; however, defining of well-balanced boundaries for storage time, staining-to-acquisition delay, and pH of washing buffer would be a valid recommendation for most applications and circumstances described herein.
Collapse
Affiliation(s)
- Łukasz Sędek
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice (SUM), 41-808 Zabrze, Poland;
| | - Juan Flores-Montero
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca (USAL), 37007 Salamanca, Spain; (J.F.-M.); (J.J.M.v.D.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Center of Biomedical Network Research in Cancer (CIBER ONC), Carlos III Institute of Health, 28029 Madrid, Spain
| | - Alita van der Sluijs
- Department of Immunology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands;
| | - Jan Kulis
- Department of Pediatric Hematology and Oncology, Medical University of Silesia in Katowice (SUM), 41-800 Zabrze, Poland; (J.K.); (T.S.)
| | - Jeroen te Marvelde
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (J.t.M.); (V.H.J.v.d.V.)
| | - Jan Philippé
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Sebastian Böttcher
- Special Hematology Laboratory, Medical Clinic III, Hematology, Oncology and Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Marieke Bitter
- European Scientific Foundation for Laboratory Hemato Oncology (ESLHO), 2333 ZA Leiden, The Netherlands;
| | - Joana Caetano
- Clinical Flow, Hemato-Oncology Unit, Champalimaud Foundation, 1400-038 Lisboa, Portugal;
| | - Vincent H. J. van der Velden
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (J.t.M.); (V.H.J.v.d.V.)
| | - Edwin Sonneveld
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
| | - Chiara Buracchi
- Pediatric Clinic of Milano-Bicocca, Tettamanti Research Center, Monza (TRC), 20900 Monza, Italy;
| | - Ana Helena Santos
- Department of Hematology, Central Hospital of Porto (CHP), 4099-001 Porto, Portugal; (A.H.S.); (M.L.)
| | - Margarida Lima
- Department of Hematology, Central Hospital of Porto (CHP), 4099-001 Porto, Portugal; (A.H.S.); (M.L.)
| | - Tomasz Szczepański
- Department of Pediatric Hematology and Oncology, Medical University of Silesia in Katowice (SUM), 41-800 Zabrze, Poland; (J.K.); (T.S.)
| | - Jacques J. M. van Dongen
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca (USAL), 37007 Salamanca, Spain; (J.F.-M.); (J.J.M.v.D.)
- Department of Immunology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands;
| | - Alberto Orfao
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca (USAL), 37007 Salamanca, Spain; (J.F.-M.); (J.J.M.v.D.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Center of Biomedical Network Research in Cancer (CIBER ONC), Carlos III Institute of Health, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-923-294-811
| |
Collapse
|
12
|
Prolonged activation of nasal immune cell populations and development of tissue-resident SARS-CoV-2-specific CD8 + T cell responses following COVID-19. Nat Immunol 2021; 23:23-32. [PMID: 34937933 DOI: 10.1038/s41590-021-01095-w] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022]
Abstract
Systemic immune cell dynamics during coronavirus disease 2019 (COVID-19) are extensively documented, but these are less well studied in the (upper) respiratory tract, where severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replicates1-6. Here, we characterized nasal and systemic immune cells in individuals with COVID-19 who were hospitalized or convalescent and compared the immune cells to those seen in healthy donors. We observed increased nasal granulocytes, monocytes, CD11c+ natural killer (NK) cells and CD4+ T effector cells during acute COVID-19. The mucosal proinflammatory populations positively associated with peripheral blood human leukocyte antigen (HLA)-DRlow monocytes, CD38+PD1+CD4+ T effector (Teff) cells and plasmablasts. However, there was no general lymphopenia in nasal mucosa, unlike in peripheral blood. Moreover, nasal neutrophils negatively associated with oxygen saturation levels in blood. Following convalescence, nasal immune cells mostly normalized, except for CD127+ granulocytes and CD38+CD8+ tissue-resident memory T cells (TRM). SARS-CoV-2-specific CD8+ T cells persisted at least 2 months after viral clearance in the nasal mucosa, indicating that COVID-19 has both transient and long-term effects on upper respiratory tract immune responses.
Collapse
|
13
|
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
|
14
|
Espasa A, Torrents S, Morales‐Indiano C, Rico LG, Bardina J, Ancochea A, Bistué‐Rovira À, Linio R, Raya M, Vergara S, Juncà J, Grifols J, Petriz J, Soria M, Sorigue M. Diagnostic performance of the ClearLLab 10C B cell tube. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:519-530. [DOI: 10.1002/cyto.b.21955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Andrea Espasa
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
| | | | - Cristian Morales‐Indiano
- Clinical Laboratory ICS‐Metropolitana Nord, Core‐Hematology Department Hospital Germans Trias i Pujol Badalona Spain
| | - Laura G. Rico
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | - Jorge Bardina
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | - Agueda Ancochea
- Banc de Sang i Teixits Hospital Germans Trias i Pujol Badalona Spain
| | - Àngel Bistué‐Rovira
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | - Rosa Linio
- Banc de Sang i Teixits Hospital Germans Trias i Pujol Badalona Spain
| | - Minerva Raya
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
| | - Sara Vergara
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
| | - Jordi Juncà
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | | | - Jordi Petriz
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | | | - Marc Sorigue
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
| |
Collapse
|
15
|
Kalina T, Bakardjieva M, Blom M, Perez-Andres M, Barendregt B, Kanderová V, Bonroy C, Philippé J, Blanco E, Pico-Knijnenburg I, Paping JHMP, Wolska-Kuśnierz B, Pac M, Tkazcyk J, Haerynck F, Akar HH, Formánková R, Freiberger T, Svatoň M, Šedivá A, Arriba-Méndez S, Orfao A, van Dongen JJM, van der Burg M. EuroFlow Standardized Approach to Diagnostic Immunopheneotyping of Severe PID in Newborns and Young Children. Front Immunol 2020; 11:371. [PMID: 32265901 PMCID: PMC7096355 DOI: 10.3389/fimmu.2020.00371] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/17/2020] [Indexed: 12/13/2022] Open
Abstract
The EuroFlow PID consortium developed a set of flow cytometry tests for evaluation of patients with suspicion of primary immunodeficiency (PID). In this technical report we evaluate the performance of the SCID-RTE tube that explores the presence of recent thymic emigrants (RTE) together with T-cell activation status and maturation stages and discuss its applicability in the context of the broader EuroFlow PID flow cytometry testing algorithm for diagnostic orientation of PID of the lymphoid system. We have analyzed peripheral blood cells of 26 patients diagnosed between birth and 2 years of age with a genetically defined primary immunodeficiency disorder: 15 severe combined immunodeficiency (SCID) patients had disease-causing mutations in RAG1 or RAG2 (n = 4, two of them presented with Omenn syndrome), IL2RG (n = 4, one of them with confirmed maternal engraftment), NHEJ1 (n = 1), CD3E (n = 1), ADA (n = 1), JAK3 (n = 3, two of them with maternal engraftment) and DCLRE1C (n = 1) and 11 other PID patients had diverse molecular defects [ZAP70 (n = 1), WAS (n = 2), PNP (n = 1), FOXP3 (n = 1), del22q11.2 (DiGeorge n = 4), CDC42 (n = 1) and FAS (n = 1)]. In addition, 44 healthy controls in the same age group were analyzed using the SCID-RTE tube in four EuroFlow laboratories using a standardized 8-color approach. RTE were defined as CD62L+CD45RO-HLA-DR-CD31+ and the activation status was assessed by the expression of HLA-DR+. Naïve CD8+ T-lymphocytes and naïve CD4+ T-lymphocytes were defined as CD62L+CD45RO-HLA-DR-. With the SCID-RTE tube, we identified patients with PID by low levels or absence of RTE in comparison to controls as well as low levels of naïve CD4+ and naïve CD8+ lymphocytes. These parameters yielded 100% sensitivity for SCID. All SCID patients had absence of RTE, including the patients with confirmed maternal engraftment or oligoclonally expanded T-cells characteristic for Omenn syndrome. Another dominant finding was the increased numbers of activated CD4+HLA-DR+ and CD8+HLA-DR+ lymphocytes. Therefore, the EuroFlow SCID-RTE tube together with the previously published PIDOT tube form a sensitive and complete cytometric diagnostic test suitable for patients suspected of severe PID (SCID or CID) as well as for children identified via newborn screening programs for SCID with low or absent T-cell receptor excision circles (TRECs).
Collapse
Affiliation(s)
- Tomas Kalina
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Marina Bakardjieva
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Maartje Blom
- Laboratory for Immunology, Department of Pediatrics, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Martin Perez-Andres
- Department of Medicine-Serv. Cytometry, Cancer Research Center (IBMCC-CSIC/USAL), University of Salamanca, Salamanca, Spain
| | - Barbara Barendregt
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Veronika Kanderová
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium.,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
| | - Elena Blanco
- Department of Medicine-Serv. Cytometry, Cancer Research Center (IBMCC-CSIC/USAL), University of Salamanca, Salamanca, Spain
| | - Ingrid Pico-Knijnenburg
- Laboratory for Immunology, Department of Pediatrics, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jitse H M P Paping
- Laboratory for Immunology, Department of Pediatrics, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | - Malgorzata Pac
- Department of Immunology, Children's Memorial Health Institute, Warsaw, Poland
| | - Jakub Tkazcyk
- Department of Pediatrics, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Filomeen Haerynck
- PID Research Lab, Department of Pediatric Pulmonology and Immunology, Ghent University Hospital, Ghent, Belgium
| | - Himmet Haluk Akar
- Department of Pediatric Immunology and Allergy, Kanuni Sultan Süleyman Training and Research Hospital, Istanbul Health Sciences University, Istanbul, Turkey
| | - Renata Formánková
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Tomáš Freiberger
- Centre for Cardiovascular Surgery and Transplantation, Brno, Czechia.,Medical Faculty, Masaryk University, Brno, Czechia
| | - Michael Svatoň
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Anna Šedivá
- Department of Immunology, University Hospital Motol, Prague, Czechia
| | - Sonia Arriba-Méndez
- Servicio de Pediatría, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine-Serv. Cytometry, Cancer Research Center (IBMCC-CSIC/USAL), University of Salamanca, Salamanca, Spain
| | - Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Mirjam van der Burg
- Laboratory for Immunology, Department of Pediatrics, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
16
|
Silva SL, Fonseca M, Pereira MLM, Silva SP, Barbosa RR, Serra-Caetano A, Blanco E, Rosmaninho P, Pérez-Andrés M, Sousa AB, Raposo AASF, Gama-Carvalho M, Victorino RMM, Hammarstrom L, Sousa AE. Monozygotic Twins Concordant for Common Variable Immunodeficiency: Strikingly Similar Clinical and Immune Profile Associated With a Polygenic Burden. Front Immunol 2019; 10:2503. [PMID: 31824477 PMCID: PMC6882918 DOI: 10.3389/fimmu.2019.02503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/07/2019] [Indexed: 01/10/2023] Open
Abstract
Monozygotic twins provide a unique opportunity to better understand complex genetic diseases and the relative contribution of heritable factors in shaping the immune system throughout life. Common Variable Immunodeficiency Disorders (CVID) are primary antibody defects displaying wide phenotypic and genetic heterogeneity, with monogenic transmission accounting for only a minority of the cases. Here, we report a pair of monozygotic twins concordant for CVID without a family history of primary immunodeficiency. They featured a remarkably similar profile of clinical manifestations and immunological alterations at diagnosis (established at age 37) and along the subsequent 15 years of follow-up. Interestingly, whole-exome sequencing failed to identify a monogenic cause for CVID, but unraveled a combination of heterozygous variants, with a predicted deleterious impact. These variants were found in genes involved in relevant immunological pathways, such as JUN, PTPRC, TLR1, ICAM1, and JAK3. The potential for combinatorial effects translating into the observed disease phenotype is inferred from their roles in immune pathways, namely in T and B cell activation. The combination of these genetic variants is also likely to impose a significant constraint on environmental influences, resulting in a similar immunological phenotype in both twins, despite exposure to different living conditions. Overall, these cases stress the importance of integrating NGS data with clinical and immunological phenotypes at the single-cell level, as provided by multi-dimensional flow-cytometry, in order to understand the complex genetic landscape underlying the vast majority of patients with CVID, as well as those with other immunodeficiencies.
Collapse
Affiliation(s)
- Susana L Silva
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal.,Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Lisbon, Portugal
| | - Mariana Fonseca
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal
| | - Marcelo L M Pereira
- Faculty of Sciences, BioISI-Biosystems & Integrative Sciences Institute, University of Lisboa, Lisbon, Portugal
| | - Sara P Silva
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal.,Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Lisbon, Portugal
| | - Rita R Barbosa
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Serra-Caetano
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal
| | - Elena Blanco
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca (USAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC, Number CB16/12/00400, Institute of Health Carlos III, Madrid, Spain
| | - Pedro Rosmaninho
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal
| | - Martin Pérez-Andrés
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca (USAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC, Number CB16/12/00400, Institute of Health Carlos III, Madrid, Spain
| | - Ana Berta Sousa
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal.,Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Lisbon, Portugal
| | - Alexandre A S F Raposo
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal
| | - Margarida Gama-Carvalho
- Faculty of Sciences, BioISI-Biosystems & Integrative Sciences Institute, University of Lisboa, Lisbon, Portugal
| | - Rui M M Victorino
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal.,Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Lisbon, Portugal
| | | | - Ana E Sousa
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.,Centro de Imunodeficiências Primárias, Centro Académico de Medicina de Lisboa, Centro Hospitalar Universitário Lisboa Norte and Faculdade de Medicina da Universidade de Lisboa and Instituto de Medicina Molecular, Lisbon, Portugal
| |
Collapse
|
17
|
Edwards ESJ, Bosco JJ, Aui PM, Stirling RG, Cameron PU, Chatelier J, Hore-Lacy F, O'Hehir RE, van Zelm MC. Predominantly Antibody-Deficient Patients With Non-infectious Complications Have Reduced Naive B, Treg, Th17, and Tfh17 Cells. Front Immunol 2019; 10:2593. [PMID: 31803177 PMCID: PMC6873234 DOI: 10.3389/fimmu.2019.02593] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Patients with predominantly antibody deficiency (PAD) suffer from severe and recurrent infections that require lifelong immunoglobulin replacement and prophylactic antibiotic treatment. Disease incidence is estimated to be 1:25,000 worldwide, and up to 68% of patients develop non-infectious complications (NIC) including autoimmunity, which are difficult to treat, causing high morbidity, and early mortality. Currently, the etiology of NIC is unknown, and there are no diagnostic and prognostic markers to identify patients at risk. Objectives: To identify immune cell markers that associate with NIC in PAD patients. Methods: We developed a standardized 11-color flow cytometry panel that was utilized for in-depth analysis of B and T cells in 62 adult PAD patients and 59 age-matched controls. Results: Nine males had mutations in Bruton's tyrosine kinase (BTK) and were defined as having X-linked agammaglobulinemia. The remaining 53 patients were not genetically defined and were clinically diagnosed with agammaglobulinemia (n = 1), common variable immunodeficiency (CVID) (n = 32), hypogammaglobulinemia (n = 13), IgG subclass deficiency (n = 1), and specific polysaccharide antibody deficiency (n = 6). Of the 53, 30 (57%) had one or more NICs, 24 patients had reduced B-cell numbers, and 17 had reduced T-cell numbers. Both PAD–NIC and PAD+NIC groups had significantly reduced Ig class-switched memory B cells and naive CD4 and CD8 T-cell numbers. Naive and IgM memory B cells, Treg, Th17, and Tfh17 cells were specifically reduced in the PAD+NIC group. CD21lo B cells and Tfh cells were increased in frequencies, but not in absolute numbers in PAD+NIC. Conclusion: The previously reported increased frequencies of CD21lo B cells and Tfh cells are the indirect result of reduced naive B-cell and T-cell numbers. Hence, correct interpretation of immunophenotyping of immunodeficiencies is critically dependent on absolute cell counts. Finally, the defects in naive B- and T-cell numbers suggest a mild combined immunodeficiency in PAD patients with NIC. Together with the reductions in Th17, Treg, and Tfh17 numbers, these key differences could be utilized as biomarkers to support definitive diagnosis and to predict for disease progression.
Collapse
Affiliation(s)
- Emily S J Edwards
- Department of Immunology and Pathology, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC, Australia.,The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia
| | - Julian J Bosco
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology Service, Department of Respiratory, Allergy and Clinical Immunology (Research), Central Clinical School, The Alfred Hospital, Melbourne, VIC, Australia
| | - Pei M Aui
- Department of Immunology and Pathology, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC, Australia.,The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia
| | - Robert G Stirling
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology Service, Department of Respiratory, Allergy and Clinical Immunology (Research), Central Clinical School, The Alfred Hospital, Melbourne, VIC, Australia
| | - Paul U Cameron
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology Service, Department of Respiratory, Allergy and Clinical Immunology (Research), Central Clinical School, The Alfred Hospital, Melbourne, VIC, Australia
| | - Josh Chatelier
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology Service, Department of Respiratory, Allergy and Clinical Immunology (Research), Central Clinical School, The Alfred Hospital, Melbourne, VIC, Australia
| | - Fiona Hore-Lacy
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology Service, Department of Respiratory, Allergy and Clinical Immunology (Research), Central Clinical School, The Alfred Hospital, Melbourne, VIC, Australia
| | - Robyn E O'Hehir
- Department of Immunology and Pathology, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC, Australia.,The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology Service, Department of Respiratory, Allergy and Clinical Immunology (Research), Central Clinical School, The Alfred Hospital, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC, Australia.,The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology Service, Department of Respiratory, Allergy and Clinical Immunology (Research), Central Clinical School, The Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Marquard AN, Carlson JCT, Weissleder R. Glass Chemistry to Analyze Human Cells under Adverse Conditions. ACS OMEGA 2019; 4:11515-11521. [PMID: 31460257 PMCID: PMC6682085 DOI: 10.1021/acsomega.9b01036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/19/2019] [Indexed: 05/17/2023]
Abstract
Emerging point-of-care diagnostic tests capable of analyzing whole mammalian cells often rely on the attachment of harvested cells to glass surfaces for subsequent molecular characterization. We set out to develop and optimize a kit for the diagnosis of lymphoma in low- and middle-income countries where access to advanced healthcare testing is often absent or prone to error. Here, we optimized a process for the lyophilization of neutravidin-coated glass and cocktails of antibodies relevant to lymphoma diagnosis to establish long-term stability of reagents required for point-of-care cell capture technology. Lyophilized glass slides showed no decline in their performance compared to freshly prepared neutravidin glass and preserved capture efficiency for 5 weeks under easily attainable storage conditions. We demonstrate the successful performance of the low-cost, lyophilized kit in a cell capture assay to enable true point-of-care analyses under adverse conditions. We anticipate that the strategy can be expanded to other cancer cell types or cell-derived vesicle analysis.
Collapse
Affiliation(s)
- Angela N. Marquard
- Center for Systems
Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
| | - Jonathan C. T. Carlson
- Center for Systems
Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- MGH Cancer
Center, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- E-mail: (J.C.T.C.)
| | - Ralph Weissleder
- Center for Systems
Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department of Systems Biology, Harvard
Medical School, 200
Longwood Avenue, Boston, Massachusetts 02115, United States
- E-mail: (R.W.)
| |
Collapse
|
19
|
van Dongen JJM, van der Burg M, Kalina T, Perez-Andres M, Mejstrikova E, Vlkova M, Lopez-Granados E, Wentink M, Kienzler AK, Philippé J, Sousa AE, van Zelm MC, Blanco E, Orfao A. EuroFlow-Based Flowcytometric Diagnostic Screening and Classification of Primary Immunodeficiencies of the Lymphoid System. Front Immunol 2019; 10:1271. [PMID: 31263462 PMCID: PMC6585843 DOI: 10.3389/fimmu.2019.01271] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/17/2019] [Indexed: 12/16/2022] Open
Abstract
Guidelines for screening for primary immunodeficiencies (PID) are well-defined and several consensus diagnostic strategies have been proposed. These consensus proposals have only partially been implemented due to lack of standardization in laboratory procedures, particularly in flow cytometry. The main objectives of the EuroFlow Consortium were to innovate and thoroughly standardize the flowcytometric techniques and strategies for reliable and reproducible diagnosis and classification of PID of the lymphoid system. The proposed EuroFlow antibody panels comprise one orientation tube and seven classification tubes and corresponding databases of normal and PID samples. The 8-color 12-antibody PID Orientation tube (PIDOT) aims at identification and enumeration of the main lymphocyte and leukocyte subsets; this includes naïve pre-germinal center (GC) and antigen-experienced post-GC memory B-cells and plasmablasts. The seven additional 8(-12)-color tubes can be used according to the EuroFlow PID algorithm in parallel or subsequently to the PIDOT for more detailed analysis of B-cell and T-cell subsets to further classify PID of the lymphoid system. The Pre-GC, Post-GC, and immunoglobulin heavy chain (IgH)-isotype B-cell tubes aim at identification and enumeration of B-cell subsets for evaluation of B-cell maturation blocks and specific defects in IgH-subclass production. The severe combined immunodeficiency (SCID) tube and T-cell memory/effector subset tube aim at identification and enumeration of T-cell subsets for assessment of T-cell defects, such as SCID. In case of suspicion of antibody deficiency, PIDOT is preferably directly combined with the IgH isotype tube(s) and in case of SCID suspicion (e.g., in newborn screening programs) the PIDOT is preferably directly combined with the SCID T-cell tube. The proposed ≥8-color antibody panels and corresponding reference databases combined with the EuroFlow PID algorithm are designed to provide fast, sensitive and cost-effective flowcytometric diagnosis of PID of the lymphoid system, easily applicable in multicenter diagnostic settings world-wide.
Collapse
Affiliation(s)
- Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Mirjam van der Burg
- Department of Immunology, Erasmus MC, Rotterdam, Netherlands.,Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Tomas Kalina
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Charles University, Prague, Czechia
| | - Martin Perez-Andres
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), CB/16/12/00233, Instituto Carlos III, Madrid, Spain
| | - Ester Mejstrikova
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Charles University, Prague, Czechia
| | - Marcela Vlkova
- Institute of Clinical Immunology and Allergology, St. Anne's University Hospital Brno, Masaryk University, Brno, Czechia
| | | | | | - Anne-Kathrin Kienzler
- Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jan Philippé
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Ana E Sousa
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Menno C van Zelm
- Department of Immunology, Erasmus MC, Rotterdam, Netherlands.,Department of Immunology and Pathology, Central Clinical School, Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Elena Blanco
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), CB/16/12/00233, Instituto Carlos III, Madrid, Spain
| | - Alberto Orfao
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), CB/16/12/00233, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
20
|
van der Burg M, Kalina T, Perez-Andres M, Vlkova M, Lopez-Granados E, Blanco E, Bonroy C, Sousa AE, Kienzler AK, Wentink M, Mejstríková E, Šinkorova V, Stuchly J, van Zelm MC, Orfao A, van Dongen JJM. The EuroFlow PID Orientation Tube for Flow Cytometric Diagnostic Screening of Primary Immunodeficiencies of the Lymphoid System. Front Immunol 2019; 10:246. [PMID: 30886612 PMCID: PMC6410673 DOI: 10.3389/fimmu.2019.00246] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/29/2019] [Indexed: 11/13/2022] Open
Abstract
In the rapidly evolving field of primary immunodeficiencies (PID), the EuroFlow consortium decided to develop a PID orientation and screening tube that facilitates fast, standardized, and validated immunophenotypic diagnosis of lymphoid PID, and allows full exchange of data between centers. Our aim was to develop a tool that would be universal for all lymphoid PIDs and offer high sensitivity to identify a lymphoid PID (without a need for specificity to diagnose particular PID) and to guide and prioritize further diagnostic modalities and clinical management. The tube composition has been defined in a stepwise manner through several cycles of design-testing-evaluation-redesign in a multicenter setting. Equally important appeared to be the standardized pre-analytical procedures (sample preparation and instrument setup), analytical procedures (immunostaining and data acquisition), the software analysis (a multidimensional view based on a reference database in Infinicyt software), and data interpretation. This standardized EuroFlow concept has been tested on 250 healthy controls and 99 PID patients with defined genetic defects. In addition, an application of new EuroFlow software tools with multidimensional pattern recognition was designed with inclusion of maturation pathways in multidimensional patterns (APS plots). The major advantage of the EuroFlow approach is that data can be fully exchanged between different laboratories in any country of the world, which is especially of interest for the PID field, with generally low numbers of cases per center.
Collapse
Affiliation(s)
- Mirjam van der Burg
- Department of Immunology, Erasmus MC, Rotterdam, Netherlands.,Department of Pediatrics, Laboratory for Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Tomas Kalina
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Martin Perez-Andres
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), 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
| | - Marcela Vlkova
- Institute of Clinical Immunology and Allergology, St Anne's University Hospital, Brno, Czechia
| | | | - Elena Blanco
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), 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
| | - Carolien Bonroy
- Laboratory for Clinical Biology and Hematology, University Hospital Ghent, Ghent, Belgium
| | - Ana E Sousa
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | | | | | - Ester Mejstríková
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Vendula Šinkorova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jan Stuchly
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Alberto Orfao
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), 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
| | - Jacques J M van Dongen
- Department of Immunology, Erasmus MC, Rotterdam, Netherlands.,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
21
|
Bouriche L, Bernot D, Nivaggioni V, Arnoux I, Loosveld M. Detection of Minimal Residual Disease in B Cell Acute Lymphoblastic Leukemia Using an Eight-Color Tube with Dried Antibody Reagents. CYTOMETRY PART B-CLINICAL CYTOMETRY 2019; 96:158-163. [PMID: 30698327 DOI: 10.1002/cyto.b.21766] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/13/2018] [Accepted: 01/09/2019] [Indexed: 11/12/2022]
Abstract
BACKGROUND Flow cytometry is a powerful tool for the detection of minimal residual disease (MRD) of B cell precursor acute lymphoblastic leukemia (BCP-ALL) patients. However, the staining process and the choice of antibodies rely on laboratory expertise and may be source of variability or technical errors. Recently, Beckman Coulter commercialized a ready to use tube with dried format reagents for BCP-ALL MRD detection. The aim of this study is to evaluate the applicability of this tube and to compare it to a conventional (liquid format reagents) method. METHODS Thirty-one samples from B ALL patients were analyzed: 19 bone marrow (BM) aspirations, 10 peripheral blood (PB) samples and 2 cerebrospinal fluids at different stages of the follow-up. In addition, we tested 5 bone marrow samples mixed into non-pathological (control) bone marrow. The dried format tube included seven antibodies: CD45Kro, CD58FITC, CD34ECD, CD10PC5.5, CD19PC7, CD38AA700, CD20AA750, with possibility of additional antibodies for blast markers identified at diagnosis. For comparison, a liquid format tube was prepared, and considered as the reference. RESULTS This tube was validated for daily routine laboratory, with satisfying qualitative (MRD + or MRD-) and quantitative (MRD percentages) correlation with the reference tube. CONCLUSION With this single dried format tube, we showed interesting results for BCP-ALL MRD detection in the aim of standardization and reliable interlaboratory results. It allows accurate MRD detection including low levels (10-4), and offers possibility to increase performance (supplementary antibody) within a preestablished effective antibody panel for BCP-ALL MRD. © 2018 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Lakhdar Bouriche
- Assistance Publique Hôpitaux de Marseille, Laboratoire d'Hématologie, Hôpital de la Timone, Marseille, France
| | - Denis Bernot
- Assistance Publique Hôpitaux de Marseille, Laboratoire d'Hématologie, Hôpital de la Timone, Marseille, France
| | - Vanessa Nivaggioni
- Assistance Publique Hôpitaux de Marseille, Laboratoire d'Hématologie, Hôpital de la Timone, Marseille, France
| | - Isabelle Arnoux
- Assistance Publique Hôpitaux de Marseille, Laboratoire d'Hématologie, Hôpital de la Timone, Marseille, France
| | - Marie Loosveld
- Assistance Publique Hôpitaux de Marseille, Laboratoire d'Hématologie, Hôpital de la Timone, Marseille, France.,CNRS, INSERM, CIML, Aix Marseille University, Marseille, France
| |
Collapse
|
22
|
Immunophenotyping Using Dried and Lyophilized Reagents. Methods Mol Biol 2019; 2032:69-79. [PMID: 31522413 DOI: 10.1007/978-1-4939-9650-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Antibody reagents that are used for flow cytometry immunophenotyping have traditionally been prepared by combining individual liquid antibody conjugates into mixtures. These cocktails have limited shelf-life, and their preparation is time-consuming and prone to laboratory error. Manufacturers of these reagents, in collaboration with several clinical and research centers, have made advances in constructing dried antibody cocktails which have addressed many of the problems inherent in preparing the liquid cocktails on the lab bench. This chapter discusses methods for the use of dried reagents.
Collapse
|