1
|
Masic D, Bell H, Van Delft FW, Irving JAE. Selection of dormant cells during treatment of T-lineage lymphoblastic leukemia and CREB as a therapeutic target. Haematologica 2024; 109:2316-2320. [PMID: 38385269 PMCID: PMC11215388 DOI: 10.3324/haematol.2023.284335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/14/2024] [Indexed: 02/23/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Dino Masic
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne
| | - Hayden Bell
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne
| | - Frederik W Van Delft
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK; Department of Paediatric Haematology and Oncology, Great North Children's Hospital, Newcastle upon Tyne
| | | |
Collapse
|
2
|
Verbeek MWC, van der Velden VHJ. The Evolving Landscape of Flowcytometric Minimal Residual Disease Monitoring in B-Cell Precursor Acute Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:4881. [PMID: 38732101 PMCID: PMC11084622 DOI: 10.3390/ijms25094881] [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: 03/29/2024] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Detection of minimal residual disease (MRD) is a major independent prognostic marker in the clinical management of pediatric and adult B-cell precursor Acute Lymphoblastic Leukemia (BCP-ALL), and risk stratification nowadays heavily relies on MRD diagnostics. MRD can be detected using flow cytometry based on aberrant expression of markers (antigens) during malignant B-cell maturation. Recent advances highlight the significance of novel markers (e.g., CD58, CD81, CD304, CD73, CD66c, and CD123), improving MRD identification. Second and next-generation flow cytometry, such as the EuroFlow consortium's eight-color protocol, can achieve sensitivities down to 10-5 (comparable with the PCR-based method) if sufficient cells are acquired. The introduction of targeted therapies (especially those targeting CD19, such as blinatumomab or CAR-T19) introduces several challenges for flow cytometric MRD analysis, such as the occurrence of CD19-negative relapses. Therefore, innovative flow cytometry panels, including alternative B-cell markers (e.g., CD22 and CD24), have been designed. (Semi-)automated MRD assessment, employing machine learning algorithms and clustering tools, shows promise but does not yet allow robust and sensitive automated analysis of MRD. Future directions involve integrating artificial intelligence, further automation, and exploring multicolor spectral flow cytometry to standardize MRD assessment and enhance diagnostic and prognostic robustness of MRD diagnostics in BCP-ALL.
Collapse
Affiliation(s)
| | - Vincent H. J. van der Velden
- Laboratory for Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
3
|
Ouyang D, Ye N, Jiang Y, Wang Y, Hu L, Chao S, Yarmush M, Tuner M, Li Y, Tang B. Label-free microfluidic chip for segregation and recovery of circulating leukemia cells: clinical applications in acute myeloid leukemia. Biomed Microdevices 2023; 26:3. [PMID: 38085348 DOI: 10.1007/s10544-023-00687-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2023] [Indexed: 12/18/2023]
Abstract
We present a label-free microfluidic chip for the segregation of circulating leukemia cells (CLCs) from blood samples, with a focus on its clinical applications in Acute Myeloid Leukemia (AML). The microfluidic chip achieved an approximate capture efficiency of 92%. The study analyzed a comprehensive set of 66 blood specimens from AML patients in different disease stages, including newly diagnosed and relapsing cases, patients in complete remission, and those in partial remission. The results showed a significant difference in CLC counts between active disease stages and remission stages (p < 0.0001), with a proposed threshold of 5 CLCs to differentiate between the two. The microfluidic chip exhibited a sensitivity of 95.4% and specificity of 100% in predicting disease recurrence. Additionally, the captured CLCs were subjected to downstream molecular analysis using droplet digital PCR, allowing for the identification of genetic mutations associated with AML. Comparative analysis with bone marrow aspirate processing by FACS demonstrated the reliability and accuracy of the microfluidic chip in tracking disease burden, with highly agreement results obtained between the two methods. The non-invasive nature of the microfluidic chip and its ability to provide real-time insights into disease progression make it a promising tool for the proactive monitoring and personalized patient care of AML.
Collapse
Affiliation(s)
- Dongfang Ouyang
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
- Shriners Hospital for Children, Boston, MA, 02114, USA.
| | - Ningxin Ye
- Department of Biology, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Yue Jiang
- Medical Imaging Science, University of Manchester, Manchester, M13 9PL, UK
| | - Yiyang Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of Californiain , Los Angeles, Los Angeles, CA, 90095, USA
| | - Lina Hu
- Department of Hematology, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Shuen Chao
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- Shriners Hospital for Children, Boston, MA, 02114, USA
| | - Martin Yarmush
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- Shriners Hospital for Children, Boston, MA, 02114, USA
| | - Memet Tuner
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- Shriners Hospital for Children, Boston, MA, 02114, USA
| | - Yonghua Li
- Department of Hematology, PLA General Hospital of Southern Theater Command, Guangzhou , Guangdong, 510010, China
| | - Bin Tang
- Department of Biomedical Engineering, South University of Science and Technology, Shenzhen , Guangdong, 518055, China.
| |
Collapse
|
4
|
Chatterjee G, Patkar N. United we stand, divided we fall. Multicentre standardization of measurable residual disease assessment in acute leukaemia is the way forward. Br J Haematol 2023; 200:277-279. [PMID: 36282207 DOI: 10.1111/bjh.18533] [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: 10/13/2022] [Accepted: 10/16/2022] [Indexed: 01/21/2023]
Abstract
Assessment of measurable residual disease (MRD) using multiparameter flow cytometry in precursor B-cell acute lymphoblastic leukaemia (ALL) is routine. However, studies on the harmonization of laboratory techniques as well as on the interpretation of results are limited. Here, Ikoma-Colturato and colleagues from Brazil demonstrate multicentric standardization of B-ALL MRD using EuroFlow protocols. Commentary on: Ikoma-Colturato et al., Multicentric standardization of minimal/measurable residual disease in B-cell precursor acute lymphoblastic leukaemia using next-generation flow cytometry in a low/middle-level income country. Br J Haematol 2023;200:381-384.
Collapse
Affiliation(s)
- Gaurav Chatterjee
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| | - Nikhil Patkar
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
| |
Collapse
|
5
|
Sampathi S, Chernyavskaya Y, Haney MG, Moore LH, Snyder IA, Cox AH, Fuller BL, Taylor TJ, Yan D, Badgett TC, Blackburn JS. Nanopore sequencing of clonal IGH rearrangements in cell-free DNA as a biomarker for acute lymphoblastic leukemia. Front Oncol 2022; 12:958673. [PMID: 36591474 PMCID: PMC9795051 DOI: 10.3389/fonc.2022.958673] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Background Acute Lymphoblastic Leukemia (ALL) is the most common pediatric cancer, and patients with relapsed ALL have a poor prognosis. Detection of ALL blasts remaining at the end of treatment, or minimal residual disease (MRD), and spread of ALL into the central nervous system (CNS) have prognostic importance in ALL. Current methods to detect MRD and CNS disease in ALL rely on the presence of ALL blasts in patient samples. Cell-free DNA, or small fragments of DNA released by cancer cells into patient biofluids, has emerged as a robust and sensitive biomarker to assess cancer burden, although cfDNA analysis has not previously been applied to ALL. Methods We present a simple and rapid workflow based on NanoporeMinION sequencing of PCR amplified B cell-specific rearrangement of the (IGH) locus in cfDNA from B-ALL patient samples. A cohort of 5 pediatric B-ALL patient samples was chosen for the study based on the MRD and CNS disease status. Results Quantitation of IGH-variable sequences in cfDNA allowed us to detect clonal heterogeneity and track the response of individual B-ALL clones throughout treatment. cfDNA was detected in patient biofluids with clinical diagnoses of MRD and CNS disease, and leukemic clones could be detected even when diagnostic cell-count thresholds for MRD were not met. These data suggest that cfDNA assays may be useful in detecting the presence of ALL in the patient, even when blasts are not physically present in the biofluid sample. Conclusions The Nanopore IGH detection workflow to monitor cell-free DNA is a simple, rapid, and inexpensive assay that may ultimately serve as a valuable complement to traditional clinical diagnostic approaches for ALL.
Collapse
Affiliation(s)
- Shilpa Sampathi
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Yelena Chernyavskaya
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Meghan G. Haney
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States,Markey Cancer Center, University of Kentucky, Lexington, KY, United States,College of Medicine, University of Kentucky, Lexington, KY, United States
| | - L. Henry Moore
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States,College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Isabel A. Snyder
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Anna H. Cox
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States,College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Brittany L. Fuller
- Department of Pediatric Oncology, University of Kentucky, Lexington, KY, United States
| | - Tamara J. Taylor
- Department of Pediatric Oncology, University of Kentucky, Lexington, KY, United States
| | - Donglin Yan
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States,Department of Biostatistics, University of Kentucky, Lexington, KY, United States
| | - Tom C. Badgett
- Department of Pediatric Oncology, University of Kentucky, Lexington, KY, United States
| | - Jessica S. Blackburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States,Markey Cancer Center, University of Kentucky, Lexington, KY, United States,*Correspondence: Jessica S. Blackburn,
| |
Collapse
|
6
|
Schwinghammer C, Koopmann J, Chitadze G, Karawajew L, Brüggemann M, Eckert C. Droplet Digital PCR: A New View on Minimal Residual Disease Quantification in Acute Lymphoblastic Leukemia. J Mol Diagn 2022; 24:856-866. [PMID: 35691569 DOI: 10.1016/j.jmoldx.2022.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 03/05/2022] [Accepted: 04/06/2022] [Indexed: 11/27/2022] Open
Abstract
Real-time quantitative PCR (qPCR) using immunoglobulin/T-cell receptor gene rearrangements has been used as the gold standard for minimal residual disease (MRD) monitoring in acute lymphoblastic leukemia (ALL) for >20 years. Recently, new PCR-based technologies have emerged, such as droplet digital PCR (ddPCR), which could offer several methodologic advances for MRD monitoring. In the current work, qPCR and ddPCR were compared in an unbiased blinded prospective study (n = 88 measurements) and in a retrospective study with selected critical low positive samples (n = 65 measurements). The former included flow cytometry (Flow; n = 31 measurements) as a third MRD detection method. Published guidelines (qPCR) and the latest, revised evaluation criteria (ie, ddPCR, Flow) have been applied for data analysis. The prospective study shows that ddPCR outperforms qPCR with a significantly better quantitative limit of detection and sensitivity. The number of critical MRD estimates below quantitative limit was reduced by sixfold and by threefold in the retrospective and prospective cohorts, respectively. Furthermore, the concordance of quantitative values between ddPCR and Flow was higher than between ddPCR and qPCR, probably because ddPCR and Flow are absolute quantification methods independent of the diagnostic sample, unlike qPCR. In summary, our data highlight the advantages of ddPCR as a more precise and sensitive technology that could be used to refine response monitoring in ALL.
Collapse
Affiliation(s)
- Claudia Schwinghammer
- Department of Paediatric Oncology/Haematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Koopmann
- Department of Haematology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Guranda Chitadze
- Department of Haematology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Leonid Karawajew
- Department of Paediatric Oncology/Haematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Monika Brüggemann
- Department of Haematology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Cornelia Eckert
- Department of Paediatric Oncology/Haematology, Charité-Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
7
|
Maurer-Granofszky M, Schumich A, Buldini B, Gaipa G, Kappelmayer J, Mejstrikova E, Karawajew L, Rossi J, Suzan AÇ, Agriello E, Anastasiou-Grenzelia T, Barcala V, Barna G, Batinić D, Bourquin JP, Brüggemann M, Bukowska-Strakova K, Burnusuzov H, Carelli D, Deniz G, Dubravčić K, Feuerstein T, Gaillard MI, Galeano A, Giordano H, Gonzalez A, Groeneveld-Krentz S, Hevessy Z, Hrusak O, Iarossi MB, Jáksó P, Kloboves Prevodnik V, Kohlscheen S, Kreminska E, Maglia O, Malusardi C, Marinov N, Martin BM, Möller C, Nikulshin S, Palazzi J, Paterakis G, Popov A, Ratei R, Rodríguez C, Sajaroff EO, Sala S, Samardzija G, Sartor M, Scarparo P, Sędek Ł, Slavkovic B, Solari L, Svec P, Szczepanski T, Taparkou A, Torrebadell M, Tzanoudaki M, Varotto E, Vernitsky H, Attarbaschi A, Schrappe M, Conter V, Biondi A, Felice M, Campbell M, Kiss C, Basso G, Dworzak MN. An Extensive Quality Control and Quality Assurance (QC/QA) Program Significantly Improves Inter-Laboratory Concordance Rates of Flow-Cytometric Minimal Residual Disease Assessment in Acute Lymphoblastic Leukemia: An I-BFM-FLOW-Network Report. Cancers (Basel) 2021; 13:cancers13236148. [PMID: 34885257 PMCID: PMC8656726 DOI: 10.3390/cancers13236148] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Standardization of flow-cytometric assessment of minimal residual disease in acute lymphoid leukemia (ALL) is necessary to allow concordant multicentric application of the methodology. This is a prerequisite for internationally collaborative trials, such as the AIEOP-BFM-ALL and the ALL IC-BFM trial. We developed and applied a comprehensive training and quality control program involving a large number of international laboratories within the I-BFM consortium to complement standardization of the methodology with an educational component as well as with persistent quality control measures to allow large ALL treatment trials which use multi-laboratory FCM-MRD assessments for risk stratification of pediatric patients with ALL. Abstract Monitoring of minimal residual disease (MRD) by flow cytometry (FCM) is a powerful prognostic tool for predicting outcomes in acute lymphoblastic leukemia (ALL). To apply FCM-MRD in large, collaborative trials, dedicated laboratory staff must be educated to concordantly high levels of expertise and their performance quality should be continuously monitored. We sought to install a unique and comprehensive training and quality control (QC) program involving a large number of reference laboratories within the international Berlin-Frankfurt-Münster (I-BFM) consortium, in order to complement the standardization of the methodology with an educational component and persistent quality control measures. Our QC and quality assurance (QA) program is based on four major cornerstones: (i) a twinning maturation program, (ii) obligatory participation in external QA programs (spiked sample send around, United Kingdom National External Quality Assessment Service (UK NEQAS)), (iii) regular participation in list-mode-data (LMD) file ring trials (FCM data file send arounds), and (iv) surveys of independent data derived from trial results. We demonstrate that the training of laboratories using experienced twinning partners, along with continuous educational feedback significantly improves the performance of laboratories in detecting and quantifying MRD in pediatric ALL patients. Overall, our extensive education and quality control program improved inter-laboratory concordance rates of FCM-MRD assessments and ultimately led to a very high conformity of risk estimates in independent patient cohorts.
Collapse
Affiliation(s)
| | - Angela Schumich
- Children’s Cancer Research Institute, Medical University of Vienna, 1090 Vienna, Austria; (M.M.-G.); (A.S.)
| | - Barbara Buldini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, 35122 Padova, Italy; (B.B.); (P.S.); (E.V.); (G.B.)
| | - Giuseppe Gaipa
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, 20900 Monza, Italy; (G.G.); (O.M.); (S.S.)
| | - Janos Kappelmayer
- Department of Laboratory Medicine, University of Debrecen, 4032 Debrecen, Hungary; (J.K.); (Z.H.)
| | - Ester Mejstrikova
- Department of Paediatric Haematology and Oncology, University Hospital Motol, 150 06 Prague, Czech Republic; (E.M.); (O.H.)
| | - Leonid Karawajew
- Department of Pediatric Oncology and Hematology, Charité Berlin, 10117 Berlin, Germany; (L.K.); (S.G.-K.)
| | - Jorge Rossi
- Cellular Immunology Laboratory, Hospital de Pediatria “Dr. Juan P. Garrahan”, Buenos Aires C1245, Argentina; (J.R.); (E.O.S.)
| | - Adın Çınar Suzan
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, 34452 Istanbul, Turkey; (A.Ç.S.); (G.D.)
| | - Evangelina Agriello
- LEB Laboratorio, Servicio de Hematologia Hospital Penna, Bahia Blanca B8000, Argentina;
| | | | - Virna Barcala
- Laboratory—Flow Cytometry, Citomlab, Buenos Aires C1406AWK, Argentina;
| | - Gábor Barna
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary;
| | - Drago Batinić
- Division of Laboratory Immunology, Department of Laboratory Diagnostics, University Hospital Centre Zagreb & School of Medicine, 10000 Zagreb, Croatia; (D.B.); (K.D.)
| | - Jean-Pierre Bourquin
- Department of Oncology and Children’s Cancer Research Center, University Children’s Hospital, 8032 Zurich, Switzerland; (J.-P.B.); (C.M.)
| | - Monika Brüggemann
- Department of Hematology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany; (M.B.); (S.K.)
| | - Karolina Bukowska-Strakova
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 31-008 Krakow, Poland;
| | - Hasan Burnusuzov
- Center of Competence “PERIMED”, Department of Pediatrics, Department of Microbiology and Clinical Immunology, Medical University Plovdiv, 4002 Plovdiv, Bulgaria;
| | | | - Günnur Deniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, 34452 Istanbul, Turkey; (A.Ç.S.); (G.D.)
| | - Klara Dubravčić
- Division of Laboratory Immunology, Department of Laboratory Diagnostics, University Hospital Centre Zagreb & School of Medicine, 10000 Zagreb, Croatia; (D.B.); (K.D.)
| | - Tamar Feuerstein
- The Rina Zaizov Division of Pediatric Hematology-Oncology, Schneider’s Children’s Medical Center, Petah Tikva 4920235, Israel;
| | - Marie Isabel Gaillard
- Bioquimica, Inmunologia, Hospital de Ninos Rocardo Gutierrez, Buenos Aires C1425EFD, Argentina;
| | - Adriana Galeano
- Flow Cytometry Laboratory, FUNDALEU, Buenos Aires C1114, Argentina;
| | - Hugo Giordano
- Fundación Pérez Scremini, Pediatric Hematology-Oncology Service, Pereira Rossell Hospital, Montevideo 11600, Uruguay;
| | | | - Stefanie Groeneveld-Krentz
- Department of Pediatric Oncology and Hematology, Charité Berlin, 10117 Berlin, Germany; (L.K.); (S.G.-K.)
| | - Zsuzsanna Hevessy
- Department of Laboratory Medicine, University of Debrecen, 4032 Debrecen, Hungary; (J.K.); (Z.H.)
| | - Ondrej Hrusak
- Department of Paediatric Haematology and Oncology, University Hospital Motol, 150 06 Prague, Czech Republic; (E.M.); (O.H.)
| | - Maria Belen Iarossi
- Flow Cytometry Laboratory, Provincial Histocompatibility Reference Centre, CUCAIBA, Buenos Aires C1114, Argentina;
| | - Pál Jáksó
- Flow Cytometry Laboratory, Department of Pathology, Clinical Centre, University of Pécs, 7622 Pécs, Hungary;
| | - Veronika Kloboves Prevodnik
- Department of Cytopathology, Institute of Oncology, 1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Saskia Kohlscheen
- Department of Hematology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany; (M.B.); (S.K.)
| | - Elena Kreminska
- Clinical Laboratory Diagnostics and Metrology of NCSH “OHMATDYT”, Ministry of Heath of Ukraine, 01601 Kiev, Ukraine;
| | - Oscar Maglia
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, 20900 Monza, Italy; (G.G.); (O.M.); (S.S.)
| | - Cecilia Malusardi
- Hospital de Clinica Jose de San Martin, Buenos Aires C1120, Argentina;
| | - Neda Marinov
- PINDA, Chilean National Pediatric Oncology Group, Hospital Roberto del Rio, Universidad de Chile, Santiago 8380418, Chile; (N.M.); (M.C.)
| | | | - Claudia Möller
- Department of Oncology and Children’s Cancer Research Center, University Children’s Hospital, 8032 Zurich, Switzerland; (J.-P.B.); (C.M.)
| | - Sergey Nikulshin
- Hematopathology and Flow Cytometry Division, Children’s Clinical University Hospital, LV-1004 Riga, Latvia;
| | | | | | - Alexander Popov
- Laboratory of Leukemia Immunophenotyping, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia;
| | - Richard Ratei
- Clinic for Hematology and Tumor Immunology, HELIOS Klinikum Berlin-Buch, 13125 Berlin, Germany;
| | - Cecilia Rodríguez
- Hospital Nacional de Clínicas, Universidad Nacional de Córdoba, Cordoba X5000HUA, Argentina;
| | - Elisa Olga Sajaroff
- Cellular Immunology Laboratory, Hospital de Pediatria “Dr. Juan P. Garrahan”, Buenos Aires C1245, Argentina; (J.R.); (E.O.S.)
| | - Simona Sala
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, 20900 Monza, Italy; (G.G.); (O.M.); (S.S.)
| | - Gordana Samardzija
- Laboratory for Flow Cytometry and Immunology, Institute for Health and Protection of Mother and Child of Serbia, 11070 Belgrade, Serbia; (G.S.); (B.S.)
| | - Mary Sartor
- The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia;
| | - Pamela Scarparo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, 35122 Padova, Italy; (B.B.); (P.S.); (E.V.); (G.B.)
| | - Łukasz Sędek
- Department of Microbiology and Immunology, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Bojana Slavkovic
- Laboratory for Flow Cytometry and Immunology, Institute for Health and Protection of Mother and Child of Serbia, 11070 Belgrade, Serbia; (G.S.); (B.S.)
| | - Liliana Solari
- Servicio de Bioquimica, Hospital Posadas, Buenos Aires B1684, Argentina;
| | - Peter Svec
- National Institute of Children’s Diseases, 831 01 Bratislava, Slovakia;
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Anna Taparkou
- Department of Pediatric Oncology Hippokration General Hospital, 546 42 Thessaloniki, Greece;
| | | | - Marianna Tzanoudaki
- Department of Immunology & Histocompatibility, “Agia Sophia” Children’s Hospital, 115 27 Athens, Greece;
| | - Elena Varotto
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, 35122 Padova, Italy; (B.B.); (P.S.); (E.V.); (G.B.)
| | - Helly Vernitsky
- Hematology Lab, Sheba Medical Center, Ramat Gan 52621, Israel;
| | - Andishe Attarbaschi
- St. Anna Children’s Hospital, Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria;
| | - Martin Schrappe
- Department of Pediatrics, University Medical Center SchleswigHolstein, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany;
| | - Valentino Conter
- Clinica Pediatrica University degli Studi di Milano Biococca, Fondazione MBBM, 20900 Monza, Italy; (V.C.); (A.B.)
| | - Andrea Biondi
- Clinica Pediatrica University degli Studi di Milano Biococca, Fondazione MBBM, 20900 Monza, Italy; (V.C.); (A.B.)
| | - Marisa Felice
- Department of Hematology and Oncology, Hospital de Pediatria “Dr. Juan P. Garrahan”, Buenos Aires C1245, Argentina;
| | - Myriam Campbell
- PINDA, Chilean National Pediatric Oncology Group, Hospital Roberto del Rio, Universidad de Chile, Santiago 8380418, Chile; (N.M.); (M.C.)
| | - Csongor Kiss
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Giuseppe Basso
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, 35122 Padova, Italy; (B.B.); (P.S.); (E.V.); (G.B.)
| | - Michael N. Dworzak
- Children’s Cancer Research Institute, Medical University of Vienna, 1090 Vienna, Austria; (M.M.-G.); (A.S.)
- St. Anna Children’s Hospital, Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria;
- Correspondence: ; Tel.: +43-1-40470-4064
| | | |
Collapse
|
8
|
Tandon S, Visser R, Astwood E, Payne J, Gray J, Wheeler L, Irving J, Virgo P. Paediatric ambiguous lineage leukaemia with monocytic differentiation at diagnosis: case series and review of literature. Br J Haematol 2021; 196:e34-e39. [PMID: 34658015 DOI: 10.1111/bjh.17852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sneha Tandon
- Division of Paediatric Haematology/Oncology, Department of Paediatrics, The Royal London Hospital, Barts Health NHS Trust, London, UK
| | | | | | | | - Juliet Gray
- Division of Paediatric Haematology/Oncology, Department of Paediatrics, The Royal London Hospital, Barts Health NHS Trust, London, UK
| | | | - Julie Irving
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | | |
Collapse
|
9
|
DePriest BP, Vieira N, Bidgoli A, Paczesny S. An overview of multiplexed analyses of CAR T-cell therapies: insights and potential. Expert Rev Proteomics 2021; 18:767-780. [PMID: 34628995 PMCID: PMC8626704 DOI: 10.1080/14789450.2021.1992276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/08/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Cancer immunotherapy is a rapidly growing field with exponential advancement in engineered immune cell-based therapies. For instance, an engineered chimeric antigen receptor (CAR) can be introduced in T-cells or other immune cells and adoptively transferred to target and kill cancer cells in hematologic malignancies or solid tumors. The first CAR-T-cell (CAR-T) therapy has been developed against CD19, a B-cell marker expressed on lymphoma and lymphoblastic leukemia. To allow for personalized treatment, proteomics approaches could provide insights into biomarkers for CAR-T therapy efficacy and toxicity. AREAS COVERED We researched the most recent technology methods of biomarker evaluation used in the laboratory and clinical setting. Publications of CAR-T biomarkers were then systematically reviewed to provide a narrative of the most validated biomarkers of CAR-T efficacy and toxicity. Examples of biomarkers include CAR-T functionality and phenotype as well as interleukin-6 and other cytokines. EXPERT COMMENTARY Biomarkers of CAR-T efficacy and toxicity have been identified, but still need to be validated and standardized across institutions. Moreover, few are used in the clinical setting due to limitations in real-time technology. Expansion of biomarker research could provide better understanding of patient response and risk of life-threatening side effects with potential for improved precision medicine.
Collapse
Affiliation(s)
- Brittany Paige DePriest
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Noah Vieira
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Alan Bidgoli
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Sophie Paczesny
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
10
|
Das N, Gupta R, Gupta SK, Bakhshi S, Seth R, Kumar C, Rai S, Singh S, Prajapati VK, Gogia A, Sahoo RK, Sharma A, Kumar L. Critical evaluation of the utility of pre- and post-therapy immunophenotypes in assessment of measurable residual disease in B-ALL. Ann Hematol 2021; 100:2487-2500. [PMID: 34236495 DOI: 10.1007/s00277-021-04580-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 06/15/2021] [Indexed: 10/20/2022]
Abstract
Measurable residual disease (MRD) is an important parameter to predict outcome in B-cell acute lymphoblastic leukemia (B-ALL). Two different approaches have been used for the assessment of MRD by multiparametric flow cytometry that include the "Leukemia Associated Aberrant Immunophenotype (LAIP)" and "Difference from Normal (DFN)" approach. In this retrospective study, we analyzed 539 samples obtained from 281 patients of which 258 were paired samples and the remaining 23 samples were from post-induction time point only, to explore the utility of baseline immunophenotype (IPT) for MRD assessment. Single-tube 10-color panel was used both at diagnosis and MRD time points. Out of 281 patients, 31.67% (n = 89) were positive and 68.32% (n = 192) were negative for MRD. Among 258 paired diagnostic and follow-up samples, baseline IPT was required in only 9.31% (24/258) cases which included cases with hematogone pattern and isolated dim to negative CD10 expression patterns. Comparison of baseline IPT with post-induction MRD positive samples showed a change in expression of at least one antigen in 94.04% cases. Although the immunophenotypic change in expression of various antigens is frequent in post-induction samples of B-ALL, it does not adversely impact the MRD assessment. In conclusion, the baseline IPT is required in less than 10% of B-ALL, specifically those with hematogone pattern and/or dim to negative expression of CD10. Hence, a combination of DFN and LAIP approach is recommended for reliable MRD assessment.
Collapse
Affiliation(s)
- Nupur Das
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India.
| | - Sanjeev Kumar Gupta
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| | - Rachna Seth
- Department of Pediatrics, AIIMS, New Delhi, India
| | - Chandan Kumar
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Sandeep Rai
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Saroj Singh
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Vijay Kumar Prajapati
- Laboratory Oncology Unit, Dr B.R. Ambedkar IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Ajay Gogia
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| | - Atul Sharma
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| | - Lalit Kumar
- Department of Medical Oncology, Dr B.R. Ambedkar IRCH, AIIMS, New Delhi, India
| |
Collapse
|
11
|
Dai Q, Zhang G, Yang H, Wang Y, Ye L, Peng L, Shi R, Guo S, He J, Jiang Y. Clinical features and outcome of pediatric acute lymphoblastic leukemia with low peripheral blood blast cell count at diagnosis. Medicine (Baltimore) 2021; 100:e24518. [PMID: 33530278 PMCID: PMC7850651 DOI: 10.1097/md.0000000000024518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 01/08/2021] [Indexed: 01/05/2023] Open
Abstract
Peripheral blood (PB) blast cell count on day 8 of prednisone therapy has been considered one of the strongest predictors of outcome in children with acute lymphoblastic leukemia (ALL). However, little is known about the clinical features and prognostic impact of PB blast cell count at diagnosis in these patients. The aim of this study was to evaluate the relationship between initial PB blast cell count and clinical prognosis of pediatric ALL.The study comprised 367 patients with ALL, aged 0 to 14 years, enrolled and treated using the Chinese Children's Leukemia Group-ALL 2008 protocol between 2011 and 2015. The majority (91.6%) of patients were B-cell precursor ALL (BCP ALL), and 8.4% were T-cell ALL (T-ALL).Patients with BCP ALL in the low PB blast cell count group (<1 × 109/L) had significantly superior survival rates to those in the high count group (≥30 × 109/L). In T-ALL, the low count group showed significantly inferior survival rates compared to both the intermediate count group (1-29.9 × 109/L) and high count group. Multivariate analysis revealed that the initial white blood cell count and minimal residual disease at the end of induction therapy were independently predictive of BCP ALL outcome, while risk stratification was shown to be an independent prognostic factor for T-ALL outcome.These results indicated that low blast cell count in PB at diagnosis was associated with different clinical outcomes in patients with BCP ALL and T-ALL, although it was not an independent outcome predictor by multivariate analysis.
Collapse
Affiliation(s)
- Qingkai Dai
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Ge Zhang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Hui Yang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Yuefang Wang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Lei Ye
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Luyun Peng
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Rui Shi
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Siqi Guo
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Jiajing He
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Yongmei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Farweez BAT, Kassim NA, Abdelfataah MF, Hassan NM, Hassnien DEA, El-Sakhawy YN. Clinical impact of early minimal residual disease detection at day 15 in precursor B-childhood acute lymphoblastic leukemia: an Egyptian experience. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2020. [DOI: 10.1186/s43042-020-00065-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Chromosomal abnormalities in childhood acute lymphoblastic leukemia (ALL) are well-established prognostic markers and useful tools for minimal residual disease (MRD) assessment. This study aimed to stratify high-risk precursor B-childhood ALL (pre-B-ALL) patients according to standard prognostic factors (age and total leucocytic count), fluorescence in situ hybridization (FISH) analysis for these cytogenetic abnormalities [t (9;22) BCR/ABL, t(1;19)TCF3/PBX1, and 11q23 MLL gene rearrangement], and MRD status at day 15. Besides, we aimed to demonstrate the relation of these prognostic factors (standard and cytogenetic risk groups) to patients’ outcome at day 15 of induction therapy as well as exploring the impact of early MRD assessment during remission induction compared to other prognostic factors together with the ability to tailor investigations as needed especially in places with limited health resources without compromising the outcome. Seventy-two newly-diagnosed Egyptian children with pre-B-ALL, aged 6 months to 15.5 years, registered from February 2016 to February 2018 were included. They were treated according to the modified Children’s Oncology Group (COG) protocol. Patients were classified into (a) standard and high-risk groups according to standard prognostic factors. (b) Patients with the studied cytogenetic abnormalities and patients without the studied cytogenetic abnormalities. (c) Good outcome (negative MRD) and bad outcome (positive MRD) groups according to day 15 MRD status.
Results
The studied cytogenetic abnormalities were identified in 22.2% of patients, all of them were in the high-risk group, and 75% of them had a bad outcome (positive MRD) at day 15 of induction therapy.
Conclusion
Patients with favorable presenting features (standard risk) and undetectable MRD after 2 weeks remission induction therapy would not be in need to advanced molecular studies, while these studies should be considered for patients with high-risk presenting features and high levels of MRD after 2 weeks remission induction therapy. Therefore, this could provide a cost-effective guideline in countries suffering from financial challenges without affecting the outcome
Collapse
|
13
|
Kim M, Park CJ. Minimal Residual Disease Detection in Pediatric Acute Lymphoblastic Leukemia. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2020. [DOI: 10.15264/cpho.2020.27.2.87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Miyoung Kim
- Department of Laboratory Medicine, Hallym University Sacred Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Anyang, Korea
| | - Chan-Jeoung Park
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| |
Collapse
|
14
|
Huang A, Chen Q, Fei Y, Wang Z, Ni X, Gao L, Chen L, Chen J, Zhang W, Yang J, Wang J, Hu X. Dynamic prediction of relapse in patients with acute leukemias after allogeneic transplantation: Joint model for minimal residual disease. Int J Lab Hematol 2020; 43:84-92. [PMID: 32881394 DOI: 10.1111/ijlh.13328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/21/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Relapse remains the leading cause of treatment failure after allogeneic hematopoietic stem cell transplantation (alloHSCT) in leukemia. Numerous investigations have demonstrated that minimal residual disease (MRD) before or after alloHSCT is prognostic of relapse risk. These MRD data were collected at specific checkpoints and could not dynamically predict the relapse risk after alloHSCT, which needs serial monitoring. METHODS In the present study, we retrospectively analyzed MRD measured with multi-parameter flow cytometry in 207 acute myeloid leukemia (AML) patients (acute promyelocytic leukemia excluded), and 124 acute B lymphoblastic leukemia (ALL) patients. A three-step method based on joint model was used to build a relapse risk prediction model. RESULTS The 3-year overall survival and relapse-free survival rates of the entire cohort were 67.1% ± 2.8% and 61.6% ± 2.8%, respectively. The model included disease status before alloHSCT, acute and chronic graft-versus-host disease, and serial MRD data. The time-dependent receiver operating characteristics was used to evaluate the ability of the model. It fitted well with actual incidence of relapse. The serial MRD data collected after alloHSCT had better discrimination capabilities for recurrence prediction with the area under the curve from 0.67 to 0.91 (AML: 0.66-0.89; ALL: 0.70-0.96). CONCLUSION The joint model was able to dynamically predict relapse-free probability after alloHSCT, which would be a useful tool to provide important information to guide decision-making in the clinic and facilitate the individualized therapy.
Collapse
Affiliation(s)
- Aijie Huang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Qi Chen
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Yang Fei
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Ziwei Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Xiong Ni
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Lei Gao
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Li Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Jie Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Weiping Zhang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Jianmin Yang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Jianmin Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Xiaoxia Hu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| |
Collapse
|
15
|
A Real-world Perspective of CD123 Expression in Acute Leukemia as Promising Biomarker to Predict Treatment Outcome in B-ALL and AML. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e673-e684. [PMID: 32561191 DOI: 10.1016/j.clml.2020.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION CD123 is overexpressed in many hematologic malignancies and found to be useful in characterizing leukemic blasts of both acute myeloid leukemia (AML) and B-acute lymphoblastic leukemia (B-ALL). CD123 has been recently found to be a marker of leukemic stem cells, and its utility to measure residual disease and potential role in disease relapse is under evaluation. MATERIALS AND METHODS Herein, we have evaluated the expression of CD123 in 757 samples of acute leukemia including 479 treatment-naive and 278 follow-up samples and compared with post-induction morphologic complete remission and measurable residual disease (MRD) status. Multiparametric flow cytometry was used for assessment of CD123 expression and immunophenotypic characterization of leukemic blasts at diagnostic and MRD assessment time points. RESULTS Using variable cutoffs of 5%, 10%, and 20% to define a case as CD123-positive, expression of CD123 was observed in 75.6%, 66.2%, and 50% of AML and 88.6%, 81.8%, and 75% of B-ALL, respectively. Of 11 patients, 7 (63.63%) had mixed phenotype acute leukemia, but none of the 12 patients with T-acute lymphoblastic leukemia showed positivity for CD123. CD123 expression at diagnosis was associated with post-induction MRD-positive status in both B-ALL (P < .001) and AML (P = .001). We also evaluated the utility of CD123 as a leukemia-associated aberrant immunophenotype and found it to be useful in both patients with AML (baseline, 50.6%; follow-up, 53%) and B-ALL (baseline, 75%; follow-up, 73.07%). CONCLUSIONS In conclusion, CD123 may be considered as a cardinal marker for residual disease assessment and response evaluation in AML and B-ALL.
Collapse
|
16
|
Don MD, Lim W, Lo A, Cox B, Huang Q, Kitahara S, Lopategui J, Alkan S. Improved Recognition of Hematogones From Precursor B-Lymphoblastic Leukemia by a Single Tube Flow Cytometric Analysis. Am J Clin Pathol 2020; 153:790-798. [PMID: 32068791 DOI: 10.1093/ajcp/aqaa007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES To improve diagnostic accuracy in differentiating hematogones from leukemic blasts in cases of precursor B-lymphoblastic leukemia/lymphoma (B-ALL), particularly those that are posttreatment or after bone marrow transplant, and to provide an algorithmic approach to this diagnostic challenge. METHODS A seven-color antibody panel including CD10, CD19, CD45, CD38, CD34, CD58, and CD81 was generated to assess the feasibility of a single tube panel and provide an algorithmic approach to distinguish hematogones from B-ALL. Fifty-three cases were analyzed, and results were correlated with histology and ancillary studies. RESULTS There was a significant difference in mean fluorescent intensity (MFI) for CD81 and CD58 when comparing hematogones and B-ALL populations (P < .001). B-ALL cases had a mean (SD) MFI of 24.6 (27.5; range, 2-125) for CD81 and 135.6 (72.6; range, 48-328) for CD58. Hematogones cases had a mean (SD) MFI of 70.2 (19.2; range, 42-123) for CD81 and 38.8 (9.4; range, 23-58) for CD58. CONCLUSIONS The flow cytometry panel with the above markers and utilization of the proposed algorithmic approach provide differentiation of hematogones from B-ALL. This includes rare cases of hematogones and B-ALL overlap where additional ancillary studies are necessary.
Collapse
Affiliation(s)
- Michelle D Don
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Washington Lim
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Amanda Lo
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Brian Cox
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Qin Huang
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Sumire Kitahara
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jean Lopategui
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Serhan Alkan
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
17
|
Abstract
Acute lymphoblastic leukaemia develops in both children and adults, with a peak incidence between 1 year and 4 years. Most acute lymphoblastic leukaemia arises in healthy individuals, and predisposing factors such as inherited genetic susceptibility or environmental exposure have been identified in only a few patients. It is characterised by chromosomal abnormalities and genetic alterations involved in differentiation and proliferation of lymphoid precursor cells. Along with response to treatment, these abnormalities are important prognostic factors. Disease-risk stratification and the development of intensified chemotherapy protocols substantially improves the outcome of patients with acute lymphoblastic leukaemia, particularly in children (1-14 years), but also in adolescents and young adults (15-39 years). However, the outcome of older adults (≥40 years) and patients with relapsed or refractory acute lymphoblastic leukaemia remains poor. New immunotherapeutic strategies, such as monoclonal antibodies and chimeric antigen receptor (CAR) T cells, are being developed and over the next few years could change the options for acute lymphoblastic leukaemia treatment.
Collapse
Affiliation(s)
- Florent Malard
- Department of Clinical Hematology and Cellular Therapy, Saint-Antoine Hospital, AP-HP, Sorbonne University, Paris, France; Sorbonne University, INSERM, Saint-Antoine Research Centre, Paris, France
| | - Mohamad Mohty
- Department of Clinical Hematology and Cellular Therapy, Saint-Antoine Hospital, AP-HP, Sorbonne University, Paris, France; Sorbonne University, INSERM, Saint-Antoine Research Centre, Paris, France.
| |
Collapse
|
18
|
Panda SS, Radhakrishnan V, Ganesan P, Rajendranath R, Ganesan TS, Rajalekshmy KR, Bhola RK, Das H, Sagar TG. Flow Cytometry Based MRD and Its Impact on Survival Outcome in Children and Young Adults with ALL: A Prospective Study from a Tertiary Cancer Centre in Southern India. Indian J Hematol Blood Transfus 2020; 36:300-308. [PMID: 32425381 PMCID: PMC7229125 DOI: 10.1007/s12288-019-01228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/02/2019] [Indexed: 11/24/2022] Open
Abstract
Presence of minimal residual disease (MRD) following induction chemotherapy is a well-recognized risk factor to predict relapse in acute lymphoblastic leukemia (ALL). There is paucity of data on MRD and outcome in ALL from India. We share our experience in establishing a flow cytometry-based MRD assay for ALL with emphasis on determination of the number of patients who had MRD on day 35 of induction therapy and its correlation with outcome and other prognostic factors. We prospectively studied MRD in patients with ALL less than 25 years who achieved morphological complete remission with induction therapy. The initial series consisted of 104 patients with ALL. Ninety-two patients had bone marrow samples collected on day 35 of remission induction chemotherapy that was adequate for MRD. Strategy of monitoring MRD was based on flow cytometry using six color staining according the leukemia associated immunophenotype found at diagnosis. Data analysis was done using Fisher exact test. The median age was 8.5 years (range 0.9-22 years). Thirty-seven out of ninety-two patients (40.2%) had MRD at end of induction. MRD on day 35 was between 0.01 and 0.1% in 18.9% of patients, between 0.1 and 1% in 59.5% and more than 1% in 21.6% patients. Among the patients who had MRD, 16.7% had favourable cytogenetics, 60% had intermediate and 13.3% had high-risk cytogenetics. The presence or absence of residual leukemia by flow cytometry at day 35 was not significantly related to age (p = 1.0), male gender (p = 0.08) hyperleukocytosis (p = 0.25) or day 8 blast clearance (p = 0.21). However, T cell phenotype (p < 0.001) was significantly associated with MRD. The 5-year event free survival (EFS) for patients who had MRD versus those who did not was 69% and 61.1% respectively (p = 0.41). The 5-year overall survival (OS) for patients who had MRD versus those who did not was 72.5% and 61.1% respectively (p = 0.33). Flow cytometric techniques can be applied to monitor MRD in patients of ALL undergoing induction therapy. Our results suggest MRD correlates with certain known prognostic factors. Though the EFS and OS was lower in MRD positive patients, the results were not statistically significant probably because of the small sample size.
Collapse
Affiliation(s)
- Soumya Surath Panda
- Department of Medical Oncology, IMS and SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, Odisha India
| | | | - Prasanth Ganesan
- Departments of Medical Oncology, Cancer Institute (WIA), Adyar, Chennai, Tamil Nadu 600020 India
| | - Rejiv Rajendranath
- Departments of Medical Oncology, Cancer Institute (WIA), Adyar, Chennai, Tamil Nadu 600020 India
| | - Trivadi S. Ganesan
- Departments of Medical Oncology, Cancer Institute (WIA), Adyar, Chennai, Tamil Nadu 600020 India
| | | | - Rajesh Kumar Bhola
- Department of Medical Oncology, IMS and SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, Odisha India
| | - Hemlata Das
- Department of Medical Oncology, IMS and SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, Odisha India
| | - Tenali Gnana Sagar
- Departments of Medical Oncology, Cancer Institute (WIA), Adyar, Chennai, Tamil Nadu 600020 India
| |
Collapse
|
19
|
Doan M, Case M, Masic D, Hennig H, McQuin C, Caicedo J, Singh S, Goodman A, Wolkenhauer O, Summers HD, Jamieson D, Delft FV, Filby A, Carpenter AE, Rees P, Irving J. Label-Free Leukemia Monitoring by Computer Vision. Cytometry A 2020; 97:407-414. [PMID: 32091180 PMCID: PMC7213640 DOI: 10.1002/cyto.a.23987] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/22/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. While there are a number of well‐recognized prognostic biomarkers at diagnosis, the most powerful independent prognostic factor is the response of the leukemia to induction chemotherapy (Campana and Pui: Blood 129 (2017) 1913–1918). Given the potential for machine learning to improve precision medicine, we tested its capacity to monitor disease in children undergoing ALL treatment. Diagnostic and on‐treatment bone marrow samples were labeled with an ALL‐discriminating antibody combination and analyzed by imaging flow cytometry. Ignoring the fluorescent markers and using only features extracted from bright‐field and dark‐field cell images, a deep learning model was able to identify ALL cells at an accuracy of >88%. This antibody‐free, single cell method is cheap, quick, and could be adapted to a simple, laser‐free cytometer to allow automated, point‐of‐care testing to detect slow early responders. Adaptation to other types of leukemia is feasible, which would revolutionize residual disease monitoring. © 2020 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Minh Doan
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Marian Case
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Dino Masic
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Holger Hennig
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Department of Systems Biology & Bioinformatics, University of Rostock, Rostock, Germany
| | - Claire McQuin
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Juan Caicedo
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Allen Goodman
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Olaf Wolkenhauer
- Department of Systems Biology & Bioinformatics, University of Rostock, Rostock, Germany
| | - Huw D Summers
- College of Engineering, Swansea University, Bay Campus, Swansea, SA1 8EN, UK
| | - David Jamieson
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Frederik V Delft
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Andrew Filby
- Flow Cytometry Core Facility. Innovation, Methodology and Application Research Theme, Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Paul Rees
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,College of Engineering, Swansea University, Bay Campus, Swansea, SA1 8EN, UK
| | - Julie Irving
- Northern Institute for Cancer Research, Newcastle University, UK
| |
Collapse
|
20
|
Gudapati P, Khanka T, Chatterjee G, Ghogale S, Badrinath Y, Deshpande N, Patil J, Narula G, Shetty D, Banavali S, Patkar NV, Gujral S, Subramanian PG, Tembhare PR. CD304/neuropilin‐1 is a very useful and dependable marker for the measurable residual disease assessment of B‐cell precursor acute lymphoblastic leukemia. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 98:328-335. [DOI: 10.1002/cyto.b.21866] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/27/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Pratyusha Gudapati
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Twinkle Khanka
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Gaurav Chatterjee
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Sitaram Ghogale
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Yajamanam Badrinath
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Nilesh Deshpande
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Jagruti Patil
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Gaurav Narula
- Homi Bhabha National Institute Mumbai Maharashtra
- Department of Pediatric OncologyTata Memorial Center, Tata Memorial Hospital, Parel Mumbai India
| | - Dhanalaxmi Shetty
- Homi Bhabha National Institute Mumbai Maharashtra
- Department of Cancer Cytogenetics, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
| | - Shripad Banavali
- Homi Bhabha National Institute Mumbai Maharashtra
- Department of Pediatric OncologyTata Memorial Center, Tata Memorial Hospital, Parel Mumbai India
| | - Nikhil V. Patkar
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Sumeet Gujral
- Homi Bhabha National Institute Mumbai Maharashtra
- Hematopathology LaboratoryTata Memorial Center, Tata Memorial Hospital Mumbai India
| | - Papagudi G. Subramanian
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| | - Prashant R. Tembhare
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Navi Mumbai India
- Homi Bhabha National Institute Mumbai Maharashtra
| |
Collapse
|
21
|
Curran KJ, Margossian SP, Kernan NA, Silverman LB, Williams DA, Shukla N, Kobos R, Forlenza CJ, Steinherz P, Prockop S, Boulad F, Spitzer B, Cancio MI, Boelens JJ, Kung AL, Khakoo Y, Szenes V, Park JH, Sauter CS, Heller G, Wang X, Senechal B, O'Reilly RJ, Riviere I, Sadelain M, Brentjens RJ. Toxicity and response after CD19-specific CAR T-cell therapy in pediatric/young adult relapsed/refractory B-ALL. Blood 2019; 134:2361-2368. [PMID: 31650176 PMCID: PMC6933289 DOI: 10.1182/blood.2019001641] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/03/2019] [Indexed: 01/04/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have demonstrated clinical benefit in patients with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). We undertook a multicenter clinical trial to determine toxicity, feasibility, and response for this therapy. A total of 25 pediatric/young adult patients (age, 1-22.5 years) with R/R B-ALL were treated with 19-28z CAR T cells. Conditioning chemotherapy included high-dose (3 g/m2) cyclophosphamide (HD-Cy) for 17 patients and low-dose (≤1.5 g/m2) cyclophosphamide (LD-Cy) for 8 patients. Fifteen patients had pretreatment minimal residual disease (MRD; <5% blasts in bone marrow), and 10 patients had pretreatment morphologic evidence of disease (≥5% blasts in bone marrow). All toxicities were reversible, including severe cytokine release syndrome in 16% (4 of 25) and severe neurotoxicity in 28% (7 of 25) of patients. Treated patients were assessed for response, and, among the evaluable patients (n = 24), response and peak CAR T-cell expansion were superior in the HD-Cy/MRD cohorts, as compared with the LD-Cy/morphologic cohorts without an increase in toxicity. Our data support the safety of CD19-specific CAR T-cell therapy for R/R B-ALL. Our data also suggest that dose intensity of conditioning chemotherapy and minimal pretreatment disease burden have a positive impact on response without a negative effect on toxicity. This trial was registered at www.clinicaltrials.gov as #NCT01860937.
Collapse
MESH Headings
- Adolescent
- Adult
- Antigens, CD19/metabolism
- Child
- Child, Preschool
- Cytokine Release Syndrome/etiology
- Cytokine Release Syndrome/pathology
- Cytokine Release Syndrome/prevention & control
- Drug Resistance, Neoplasm
- Female
- Humans
- Infant
- Male
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/therapy
- Neoplasm, Residual/etiology
- Neoplasm, Residual/pathology
- Neoplasm, Residual/prevention & control
- Neurotoxicity Syndromes/etiology
- Neurotoxicity Syndromes/pathology
- Neurotoxicity Syndromes/prevention & control
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/immunology
- Salvage Therapy
- Survival Rate
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Treatment Outcome
- Young Adult
Collapse
Affiliation(s)
- Kevin J Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Steven P Margossian
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children's Hosptial, Boston, MA
| | - Nancy A Kernan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lewis B Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children's Hosptial, Boston, MA
| | - David A Williams
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children's Hosptial, Boston, MA
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rachel Kobos
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Peter Steinherz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Susan Prockop
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Farid Boulad
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Barbara Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Maria I Cancio
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Jaap Jan Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yasmin Khakoo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Victoria Szenes
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jae H Park
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Craig S Sauter
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Glenn Heller
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xiuyan Wang
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
- Michael G. Harris Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Brigitte Senechal
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
- Michael G. Harris Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Isabelle Riviere
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Michael G. Harris Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michel Sadelain
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J Brentjens
- Center For Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
22
|
Hendricks CL, Buldeo S, Pillay D, Naidoo A, Thejpal R, Rapiti N, Neethling B, Goga Y, van Staaden H. Comparing morphology, flow cytometry and molecular genetics in the assessment of minimal residual disease in children with B-acute lymphoblastic leukaemia (B-ALL). SOUTH AFRICAN JOURNAL OF ONCOLOGY 2019. [DOI: 10.4102/sajo.v3i0.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
23
|
Rocha JMC, Xavier SG, Souza MEDL, Murao M, de Oliveira BM. Comparison between flow cytometry and standard PCR in the evaluation of MRD in children with acute lymphoblastic leukemia treated with the GBTLI LLA - 2009 protocol. Pediatr Hematol Oncol 2019; 36:287-301. [PMID: 31287348 DOI: 10.1080/08880018.2019.1636168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Minimal residual disease (MRD) monitoring is of prognostic importance in childhood acute lymphoblastic leukemia (ALL). The detection of immunoglobulin and T-cell receptor gene rearrangements by real-time quantitative PCR (RT-PCR) is considered the gold standard for this evaluation. However, more accessible methods also show satisfactory performance. This study aimed to compare MRD analysis by four-color flow cytometry (FC) and qualitative standard PCR on days 35 and 78 of chemotherapy and to correlate these data with patients' clinical characteristics. Forty-two children with a recent diagnosis of ALL, admitted to a public hospital in Brazil for treatment in accordance with the Brazilian Childhood Cooperative Group for ALL Treatment (GBTLI LLA-2009), were included. Bone marrow samples collected at diagnosis and on days 35 and 78 of treatment were analyzed for the immunophenotypic characterization of blasts by FC and for the detection of clonal rearrangements by standard PCR. Paired analyses were performed in 61/68 (89.7%) follow-up samples, with a general agreement of 88.5%. Disagreements were resolved by RT-PCR, which evidenced one false-negative and four false-positive results in FC, as well as two false-negative results in PCR. Among the prognostic factors, a significant association was found only between T-cell lineage and MRD by standard PCR. These results show that FC and standard PCR produce similar results in MRD detection of childhood ALL and that both methodologies may be useful in the monitoring of disease treatment, especially in regions with limited financial resources.
Collapse
Affiliation(s)
| | | | | | - Mitiko Murao
- Federal University of Minas Gerais (UFMG) , Belo Horizonte , MG , Brazil
| | | |
Collapse
|
24
|
Reiter M, Diem M, Schumich A, Maurer-Granofszky M, Karawajew L, Rossi JG, Ratei R, Groeneveld-Krentz S, Sajaroff EO, Suhendra S, Kampel M, Dworzak MN. Automated Flow Cytometric MRD Assessment in Childhood Acute B- Lymphoblastic Leukemia Using Supervised Machine Learning. Cytometry A 2019; 95:966-975. [PMID: 31282025 DOI: 10.1002/cyto.a.23852] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/30/2019] [Accepted: 05/28/2019] [Indexed: 12/22/2022]
Abstract
Minimal residual disease (MRD) as measured by multiparameter flow cytometry (FCM) is an independent and strong prognostic factor in B-cell acute lymphoblastic leukemia (B-ALL). However, reliable flow cytometric detection of MRD strongly depends on operator skills and expert knowledge. Hence, an objective, automated tool for reliable FCM-MRD quantification, able to overcome the technical diversity and analytical subjectivity, would be most helpful. We developed a supervised machine learning approach using a combination of multiple Gaussian Mixture Models (GMM) as a parametric density model. The approach was used for finding the weights of a linear combination of multiple GMMs to represent new, "unseen" samples by an interpolation of stored samples. The experimental data set contained FCM-MRD data of 337 bone marrow samples collected at day 15 of induction therapy in three different laboratories from pediatric patients with B-ALL for which accurate, expert-set gates existed. We compared MRD quantification by our proposed GMM approach to operator assessments, its performance on data from different laboratories, as well as to other state-of-the-art automated read-out methods. Our proposed GMM-combination approach proved superior over support vector machines, deep neural networks, and a single GMM approach in terms of precision and average F 1 -scores. A high correlation of expert operator-based and automated MRD assessment was achieved with reliable automated MRD quantification (F 1 -scores >0.5 in more than 95% of samples) in the clinically relevant range. Although best performance was found, if test and training samples were from the same system (i.e., flow cytometer and staining panel; lowest median F 1 -score 0.92), cross-system performance remained high with a median F 1 -score above 0.85 in all settings. In conclusion, our proposed automated approach could potentially be used to assess FCM-MRD in B-ALL in an objective and standardized manner across different laboratories. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Michael Reiter
- Immunological Diagnostics, Children's Cancer Research Institute, Vienna, Austria.,Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Markus Diem
- Immunological Diagnostics, Children's Cancer Research Institute, Vienna, Austria.,Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Angela Schumich
- Immunological Diagnostics, Children's Cancer Research Institute, Vienna, Austria
| | | | - Leonid Karawajew
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jorge G Rossi
- Cellular Immunology Laboratory, Hospital de Pediatria "Dr. Juan P. Garrahan", Buenos Aires, Argentina
| | - Richard Ratei
- Department of Hematology, Oncology and Tumor Immunology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | | | - Elisa O Sajaroff
- Cellular Immunology Laboratory, Hospital de Pediatria "Dr. Juan P. Garrahan", Buenos Aires, Argentina
| | | | - Martin Kampel
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Michael N Dworzak
- Immunological Diagnostics, Children's Cancer Research Institute, Vienna, Austria.,Labdia Labordiagnostik GmbH, Vienna, Austria
| | | |
Collapse
|
25
|
Tembhare PR, Subramanian PG PG, Ghogale S, Chatterjee G, Patkar NV, Gupta A, Shukla R, Badrinath Y, Deshpande N, Narula G, Rodrigues P, Girase K, Dhaliwal D, Prasad M, Shetty D, Banavali S, Gujral S. A High‐Sensitivity 10‐Color Flow Cytometric Minimal Residual Disease Assay in B‐Lymphoblastic Leukemia/Lymphoma Can Easily Achieve the Sensitivity of 2‐in‐10
6
and Is Superior to Standard Minimal Residual Disease Assay: A Study of 622 Patients. CYTOMETRY PART B-CLINICAL CYTOMETRY 2019; 98:57-67. [DOI: 10.1002/cyto.b.21831] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/23/2019] [Accepted: 05/30/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Prashant R. Tembhare
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | | | - Sitaram Ghogale
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Gaurav Chatterjee
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Nikhil V. Patkar
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Avinash Gupta
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Rahul Shukla
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Yajamanam Badrinath
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Nilesh Deshpande
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Gaurav Narula
- Department of Pediatric Oncology, Tata Memorial CenterTata Memorial Hospital Mumbai Maharashtra 400012 India
| | - Pearl Rodrigues
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Karishma Girase
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Dilshad Dhaliwal
- Hematopathology Laboratory, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Maya Prasad
- Department of Pediatric Oncology, Tata Memorial CenterTata Memorial Hospital Mumbai Maharashtra 400012 India
| | - Dhanalaxmi Shetty
- Department of Cancer Cytogenetics, ACTREC, Tata Memorial CenterHBNI University Mumbai Maharashtra 410210 India
| | - Shripad Banavali
- Department of Pediatric Oncology, Tata Memorial CenterTata Memorial Hospital Mumbai Maharashtra 400012 India
| | - Sumeet Gujral
- Hematopathology LaboratoryTata Memorial Hospital, Tata Memorial Center Mumbai Maharashtra 400012 India
| |
Collapse
|
26
|
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: 2.2] [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
|
27
|
Buldini B, Maurer-Granofszky M, Varotto E, Dworzak MN. Flow-Cytometric Monitoring of Minimal Residual Disease in Pediatric Patients With Acute Myeloid Leukemia: Recent Advances and Future Strategies. Front Pediatr 2019; 7:412. [PMID: 31681710 PMCID: PMC6798174 DOI: 10.3389/fped.2019.00412] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/25/2019] [Indexed: 01/10/2023] Open
Abstract
Minimal residual disease (MRD) by multiparametric flow cytometry (MFC) has been recently shown as a strong and independent prognostic marker of relapse in pediatric AML (pedAML) when measured at specific time points during Induction and/or Consolidation therapy. Hence, MFC-MRD has the potential to refine the current strategies of pedAML risk stratification, traditionally based on the cytogenetic and molecular genetic aberrations at diagnosis. Consequently, it may guide the modulation of therapy intensity and clinical decision making. However, the use of non-standardized protocols, including different staining panels, analysis, and gating strategies, may hamper a broad implementation of MFC-MRD monitoring in clinical routine. Besides, the thresholds of MRD positivity still need to be validated in large, prospective and multi-center clinical studies, as well as optimal time points of MRD assessment during therapy, to better discriminate patients with different prognosis. In the present review, we summarize the most relevant findings on MFC-MRD testing in pedAML. We examine the clinical significance of MFC-MRD and the recent advances in its standardization, including innovative approaches with an automated analysis of MFC-MRD data. We also touch upon other technologies for MRD assessment in AML, such as quantitative genomic breakpoint PCR, current challenges and future strategies to enable full incorporation of MFC-MRD into clinical practice.
Collapse
Affiliation(s)
- Barbara Buldini
- Laboratory of Hematology-Oncology, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | | | - Elena Varotto
- Laboratory of Hematology-Oncology, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | - Michael N Dworzak
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Vienna, Austria
| |
Collapse
|
28
|
Zhao X, Zhao X, Chen H, Qin Y, Xu L, Zhang X, Liu K, Huang X, Chang YJ. Comparative Analysis of Flow Cytometry and RQ-PCR for the Detection of Minimal Residual Disease in Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2018; 24:1936-1943. [DOI: 10.1016/j.bbmt.2018.03.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/12/2018] [Indexed: 01/01/2023]
|
29
|
Gaipa G, Buracchi C, Biondi A. Flow cytometry for minimal residual disease testing in acute leukemia: opportunities and challenges. Expert Rev Mol Diagn 2018; 18:775-787. [DOI: 10.1080/14737159.2018.1504680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Giuseppe Gaipa
- Department of Pediatrics, University of Milano-Bicocca, Fondazione Tettamanti - Centro Ricerca M.Tettamanti, Monza, Italy
| | - Chiara Buracchi
- Department of Pediatrics, University of Milano-Bicocca, Fondazione Tettamanti - Centro Ricerca M.Tettamanti, Monza, Italy
| | - A Biondi
- Department of Pediatrics, University of Milano-Bicocca, Fondazione Tettamanti - Centro Ricerca M.Tettamanti, Monza, Italy
- Fondazione MBBM/Ospedale San Gerardo - Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
30
|
How to make usage of the standardized EuroFlow 8-color protocols possible for instruments of different manufacturers. J Immunol Methods 2017; 475:112388. [PMID: 29154914 DOI: 10.1016/j.jim.2017.11.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 11/14/2017] [Indexed: 11/21/2022]
Abstract
A critical component of the EuroFlow standardization of leukemia/lymphoma immunophenotyping is instrument setup. Initially, the EuroFlow consortium developed a step-by-step standard operating protocol for instrument setup of ≥8-color flow cytometers that were available in 2006, when the EuroFlow activities started. Currently, there are 14 instruments from 9 manufacturers capable of 3-laser excitation and ≥8 color measurements. The specific adaptations required in the instrument set-up to enable them to acquire the standardized 8-color EuroFlow protocols are described here. Overall, all 14 instruments can be fitted with similar violet, blue and red lasers for simultaneous measurements of ≥8 fluorescent dyes. Since individual instruments differ both on their dynamic range (scale) and emission filters, it is not accurate to simply recalculate the target values to different scale, but adjustment of PMT voltages to a given emission filter and fluorochrome, is essential. For this purpose, EuroFlow has developed an approach using Type IIB (spectrally matching) particles to set-up standardized and fully comparable fluorescence measurements, in instruments from different manufacturers, as demonstrated here for the FACSCanto II, and Navios and MACSQuant flow cytometers. Data acquired after such adjustment on any of the tested cytometry platforms could be fully superimposed and therefore analyzed together. The proposed approach can be used to derive target values for any combination of spectrally distinct fluorochromes and any distinct emission filter of any new flow cytometry platform, which enables the measurement of the 8-color EuroFlow panels in a standardized way, by creating superimposable datafiles.
Collapse
|
31
|
Muccio VE, Saraci E, Gilestro M, Oddolo D, Ruggeri M, Caltagirone S, Bruno B, Boccadoro M, Omedè P. Relevance of sample preparation for flow cytometry. Int J Lab Hematol 2017; 40:152-158. [DOI: 10.1111/ijlh.12755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 09/04/2017] [Indexed: 11/26/2022]
Affiliation(s)
- V. E. Muccio
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| | - E. Saraci
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| | - M. Gilestro
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| | - D. Oddolo
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| | - M. Ruggeri
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| | - S. Caltagirone
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| | - B. Bruno
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| | - M. Boccadoro
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| | - P. Omedè
- Divisione Universitaria di Ematologia; A.O.U. Città della Salute e della Scienza di Torino; Torino Italy
| |
Collapse
|
32
|
Chatterjee G, Gujral S, Subramanian PG, Tembhare PR. Clinical Relevance of Multicolour Flow Cytometry in Plasma Cell Disorders. Indian J Hematol Blood Transfus 2017; 33:303-315. [PMID: 28824230 PMCID: PMC5544653 DOI: 10.1007/s12288-017-0822-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/25/2017] [Indexed: 01/06/2023] Open
Abstract
Multicolor flow cytometric (MFC) immunophenotyping is one of the basic test that is needed in the evaluation of hematolymphoid malignancies. Previously, there has been some reluctance in the use of MFC in plasma cell disorders (PCD). It was mainly due tolack of standardization, inadequate experience and detection of the lower number of plasma cells by MFC as compared to morphology. However, MFC has gone through many technological advancements in the last few years and a wide variety of reagents are now commercially available which worldwide allowed the establishment of standardized sensitive MFC-based immunophenotypic assay for PCD. Various studies have proven that MFC has a high clinical relevance in the diagnosis and risk stratification of multiple myeloma, its precursor conditions and other PCDs. Moreover, recent studies have shown that MFC is a highly sensitive and reliable technique for the monitoring of clinical response in the era of novel therapies. In this review, we have discussed the various applications of MFC in the management of PCD and their clinical relevance.
Collapse
Affiliation(s)
- Gaurav Chatterjee
- Hematopathology Laboratory, Tata Memorial Center, Room 17-18, CCE Building, ACTREC, Tata Memorial Center, Kharghar, Navi Mumbai, 410210 Maharashtra India
| | - Sumeet Gujral
- Hematopathology Laboratory, Tata Memorial Center, Room 17-18, CCE Building, ACTREC, Tata Memorial Center, Kharghar, Navi Mumbai, 410210 Maharashtra India
| | - Papagudi G. Subramanian
- Hematopathology Laboratory, Tata Memorial Center, Room 17-18, CCE Building, ACTREC, Tata Memorial Center, Kharghar, Navi Mumbai, 410210 Maharashtra India
| | - Prashant R. Tembhare
- Hematopathology Laboratory, Tata Memorial Center, Room 17-18, CCE Building, ACTREC, Tata Memorial Center, Kharghar, Navi Mumbai, 410210 Maharashtra India
| |
Collapse
|
33
|
Berry DA, Zhou S, Higley H, Mukundan L, Fu S, Reaman GH, Wood BL, Kelloff GJ, Jessup JM, Radich JP. Association of Minimal Residual Disease With Clinical Outcome in Pediatric and Adult Acute Lymphoblastic Leukemia: A Meta-analysis. JAMA Oncol 2017; 3:e170580. [PMID: 28494052 DOI: 10.1001/jamaoncol.2017.0580] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Minimal residual disease (MRD) refers to the presence of disease in cases deemed to be in complete remission by conventional pathologic analysis. Assessing the association of MRD status following induction therapy in patients with acute lymphoblastic leukemia (ALL) with relapse and mortality may improve the efficiency of clinical trials and accelerate drug development. Objective To quantify the relationships between event-free survival (EFS) and overall survival (OS) with MRD status in pediatric and adult ALL using publications of clinical trials and other databases. Data Sources Clinical studies in ALL identified via searches of PubMed, MEDLINE, and clinicaltrials.gov. Study Selection Our search and study screening process adhered to the PRISMA Guidelines. Studies that addressed EFS or OS by MRD status in patients with ALL were included; reviews, abstracts, and studies with fewer than 30 patients or insufficient MRD description were excluded. Data Extraction and Synthesis Study sample size, patient age, follow-up time, timing of MRD assessment (postinduction or consolidation), MRD detection method, phenotype/genotype (B cell, T cell, Philadelphia chromosome), and EFS and OS. Searches of PubMed and MEDLINE identified 566 articles. A parallel search on clinicaltrials.gov found 67 closed trials and 62 open trials as of 2014. Merging results of 2 independent searches and applying exclusions gave 39 publications in 3 arms of patient populations (adult, pediatric, and mixed). We performed separate meta-analyses for each of these 3 subpopulations. Results The 39 publications comprised 13 637 patients: 16 adult studies (2076 patients), 20 pediatric (11 249 patients), and 3 mixed (312 patients). The EFS hazard ratio (HR) for achieving MRD negativity is 0.23 (95% Bayesian credible interval [BCI] 0.18-0.28) for pediatric patients and 0.28 (95% BCI, 0.24-0.33) for adults. The respective HRs in OS are 0.28 (95% BCI, 0.19-0.41) and 0.28 (95% BCI, 0.20-0.39). The effect was similar across all subgroups and covariates. Conclusions and Relevance The value of having achieved MRD negativity is substantial in both pediatric and adult patients with ALL. These results are consistent across therapies, methods of and times of MRD assessment, cutoff levels, and disease subtypes. Minimal residual disease status warrants consideration as an early measure of disease response for evaluating new therapies, improving the efficiency of clinical trials, accelerating drug development, and for regulatory approval. A caveat is that an accelerated approval of a particular new drug using an intermediate end point, such as MRD, would require confirmation using traditional efficacy end points.
Collapse
Affiliation(s)
- Donald A Berry
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston
| | | | | | - Shuangshuang Fu
- University of Texas Health Science Center at Houston, Houston
| | | | - Brent L Wood
- University of Washington School of Medicine, St Louis, Missouri
| | | | | | | |
Collapse
|
34
|
Keeney M, Wood BL, Hedley BD, DiGiuseppe JA, Stetler-Stevenson M, Paietta E, Lozanski G, Seegmiller AC, Greig BW, Shaver AC, Mukundan L, Higley HR, Sigman CC, Kelloff G, Jessup JM, Borowitz MJ. A QA Program for MRD Testing Demonstrates That Systematic Education Can Reduce Discordance Among Experienced Interpreters. CYTOMETRY PART B-CLINICAL CYTOMETRY 2017; 94:239-249. [PMID: 28475275 DOI: 10.1002/cyto.b.21528] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/30/2017] [Accepted: 04/10/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Minimal residual disease (MRD) in B lymphoblastic leukemia (B-ALL) by flow cytometry is an established prognostic factor used to adjust treatment in most pediatric therapeutic protocols. MRD in B-ALL has been standardized by the Children's Oncology Group (COG) in North America, but not routine clinical labs. The Foundation for National Institutes of Health sought to harmonize MRD measurement among COG, oncology groups, academic, community and government, laboratories. METHODS Listmode data from post-induction marrows were distributed from a reference lab to seven different clinical FCM labs with variable experience in B-ALL MRD. Labs were provided with the COG protocol. Files from 15 cases were distributed to the seven labs. Educational sessions were implemented, and 10 more listmode file cases analyzed. RESULTS Among 105 initial challenges, the overall discordance rate was 26%. In the final round, performance improved considerably; out of 70 challenges, there were five false positives and one false negative (9% discordance), and no quantitative discordance. Four of six deviations occurred in a single lab. Three samples with hematogones were still misclassified as MRD. CONCLUSIONS Despite the provision of the COG standardized analysis protocol, even experienced laboratories require an educational component for B-ALL MRD analysis by FCM. Recognition of hematogones remains challenging for some labs when using the COG protocol. The results from this study suggest that dissemination of MRD testing to other North American laboratories as part of routine clinical management of B-ALL is possible but requires additional educational components to complement standardized methodology. © 2017 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Michael Keeney
- Pathology and Laboratory Medicine, London Health Sciences Centre, London, Ontario, Canada
| | - Brent L Wood
- Seattle Cancer Care Alliance, Seattle, Washington.,University of Washington, Seattle, Washington
| | - Benjamin D Hedley
- Pathology and Laboratory Medicine, London Health Sciences Centre, London, Ontario, Canada
| | | | | | | | - Gerard Lozanski
- Department of Pathology, Ohio State University, Columbus, Ohio
| | - Adam C Seegmiller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bruce W Greig
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Aaron C Shaver
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | - Gary Kelloff
- Cancer Imaging Program, National Cancer Institute, Bethesda, Maryland
| | | | - Michael J Borowitz
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
35
|
Snowden JA, O'Connell S, Hawkins J, Dalley C, Jack A, Mannari D, McNamara C, Scott M, Shenton G, Soilleux E, Macbeth F. Haematological cancers: improving outcomes. A summary of updated NICE service guidance in relation to Specialist Integrated Haematological Malignancy Diagnostic Services (SIHMDS). J Clin Pathol 2017; 70:461-468. [PMID: 28389440 DOI: 10.1136/jclinpath-2016-204029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 11/04/2022]
Abstract
Haematological malignancies are a diverse group of cancers that affect the blood, bone marrow and lymphatic systems. Laboratory diagnosis of haematological malignancies is dependent on combining several technologies, including morphology, immunophenotyping, cytogenetics and molecular genetics correlated clinical details and classification according to the current WHO guidelines. The concept of the Specialised Integrated Haematological Malignancy Diagnostic Services (SIHMDS) has evolved since the UK National Institute for Health and Care Excellence (NICE) Improving Outcomes Guidance (IOG) in 2003 and subsequently various models of delivery have been established. As part of the 2016 update to the NICE IOG, these models were systematically evaluated and recommendations produced to form the basis for quality standards for future development of SIHMDS. We provide a summary of the systematic review and recommendations. Although the recommendations pertain to the UK National Health Service (NHS), they have relevance to the modern delivery of diagnostic services internationally.
Collapse
Affiliation(s)
- John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Susan O'Connell
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - James Hawkins
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Chris Dalley
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Andrew Jack
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Deepak Mannari
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Chris McNamara
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Mike Scott
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Geoff Shenton
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Elizabeth Soilleux
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Fergus Macbeth
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | | |
Collapse
|
36
|
Baraka A, Sherief LM, Kamal NM, Shorbagy SE. Detection of minimal residual disease in childhood B-acute lymphoblastic leukemia by 4-color flowcytometry. Int J Hematol 2017; 105:784-791. [PMID: 28324281 DOI: 10.1007/s12185-017-2206-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 11/28/2022]
Abstract
Monitoring of minimal residual disease (MRD) is currently considered the most powerful predictor of outcome in acute lymphoblastic leukemia (ALL). Achievement of a negative MRD state assessed by multicolor flowcytometry (MFC) is an important predictor of disease-free survival (DFS) and overall survival (OS) in ALL patients. We sought to determine whether panels of antibodies combination are more suitable for detection of MRD in Childhood ALL. Eighty-four (84) patients with ALL (B-lineage subtype) were enrolled in this study. Normal template for B cell precursors was established in 15 control participants using 4-four panels of monoclonal Antibodies (Mo Abs),{CD22, CD45, CD58 and CD97 in combination with CD10, CD19, CD34}. At diagnosis, CD22 exhibited the lowest incidence of expression in only 50% of all patients, while CD45, CD58, and CD97 were expressed in 80.9, 59.5 and 92.8%, respectively. Analysis of MRD was performed for each Mo Abs combination at day 0 and day 14 post-induction of chemotherapy by 4-color (FCM). The incidence of MRD was 61.9, 70.6, 60.0 and 55.1% for CD22, CD45, CD58 and CD97, respectively. In B-ALL patients, (CD10/CD19/CD34/CD45) + (CD10/CD19/CD34/CD97) represented the highest incidence of expression of leukemic cells markers with a significant correlation with blasts count, suggesting that these are more specific for MRD detection. Also FCM is relatively cost effective for detection of MRD in ALL patients and its applicability in routine leukemia lab is valuable. MRD evaluation at the end of the induction therapy (i.e. day 35 or 42 according to the different schedules) is advised. Also, Ig/T cell receptor gene rearrangements and gene fusions analyzed by polymerase chain reaction (PCR) are preferred.
Collapse
Affiliation(s)
- Ahmad Baraka
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Laila M Sherief
- Department of Pediatric, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Naglaa M Kamal
- Department of Pediatric, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Shereen El Shorbagy
- Department of Medical Oncology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
37
|
Burnusuzov HA, Spasova MI, Murdjeva MA, Stoyanova AA, Mumdziev IN, Kaleva VI, Belcheva MI, Bosheva MN. Immunophenotypic Modulation of the Blast Cells in Childhood Acute Lymphoblastic Leukemia Minimal Residual Disease Detection. Folia Med (Plovdiv) 2017; 58:28-35. [PMID: 27383875 DOI: 10.1515/folmed-2016-0004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/11/2016] [Indexed: 11/15/2022] Open
Abstract
UNLABELLED Early clearance of leukemic cells during induction therapy of childhood acute lymphoblastic leukemia (ALL) is a basis for treatment optimization. Currently, the most widely used methods for the detection of minute residual malignant cells in the bone marrow and/or peripheral blood, minimal residual disease (MRD), are PCR and flow cytometry (FCM). Immunophenotypic modulation (IM) is a well known factor that can hamper the accurate FCM analysis. AIM To report the IM detected by 8-color FCM during the BFM-type remission induction in 24 consecutive MRD-positive samples of children with B-cell precursor ALL and the possible implications for MRD detection. PATIENTS AND METHODS Between 2010 and 2012 we prospectively followed up the MRD on days 15 and 33 of induction treatment in bone marrow (BM) samples and on day 8 in peripheral blood (PB). The IM was assessed by comparative analyses of the changes in the mean fluorescence intensity of 7 highly relevant antigens expressed by the leukemic cells and normal B-lymphocytes. RESULTS IM occurred, to different extents, in all analyzed day 15 BM and in most day 33 BM samples. Statistically significant changes in the MFI-levels of four CDs expressed by the leukemic blasts were observed: downmodulation of CD10, CD19 and CD34 and upmodulation of CD20. No changes in the expression of CD38, CD58 and CD45 were noticed. CONCLUSIONS Measuring the MRD by standardized 8-color flow cytometry helps improve the monitoring of the disease, leading to better therapeutic results. However, the IM of the different antigens expressed by the leukemic blasts should be taken into consideration and cautiously analyzed.
Collapse
Affiliation(s)
- Hasan A Burnusuzov
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Mariya I Spasova
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Mariana A Murdjeva
- Department of Microbiology and Immunology, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Angelina A Stoyanova
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Ivan N Mumdziev
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Valeriya I Kaleva
- Department of Pediatrics and Medical Genetics, Medical University of Varna, Varna, Bulgaria
| | - Milena I Belcheva
- Department of Pediatrics and Medical Genetics, Medical University of Varna, Varna, Bulgaria
| | - Miroslava N Bosheva
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
38
|
Sala Torra O, Othus M, Williamson DW, Wood B, Kirsch I, Robins H, Beppu L, O'Donnell MR, Forman SJ, Appelbaum FR, Radich JP. Next-Generation Sequencing in Adult B Cell Acute Lymphoblastic Leukemia Patients. Biol Blood Marrow Transplant 2017; 23:691-696. [PMID: 28062215 DOI: 10.1016/j.bbmt.2016.12.639] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/28/2016] [Indexed: 12/16/2022]
Abstract
We used next-generation sequencing (NGS) of the immunoglobulin genes to evaluate residual disease in 153 specimens from 32 patients with adult B cell acute lymphoblastic leukemia enrolled in a single multicenter study. The sequencing results were compared with multiparameter flow cytometry (MFC) data in 66 specimens (25 patients) analyzed by both methods. There was a strong concordance (82%) between the methods in the qualitative determination of the presence of disease. However, in 17% of cases, leukemia was detected by sequencing but not by MFC. In 54 bone marrow (BM) and peripheral blood (PB) paired specimens, the burden of leukemia detected by NGS was lower in PB than in BM, although it was still detectable in 68% of the 28 paired specimens with positive BM. Lastly, patients without disease detected by NGS or MFC had a 5-year relapse free survival of > 80%. The results suggest that residual disease detection by immunoglobulin gene sequencing is an extremely sensitive technique and may identify patients that might benefit from transplantation. Moreover, the increased sensitivity of the method may allow frequent peripheral blood testing to supplement marrow sampling to measure disease response.
Collapse
Affiliation(s)
- Olga Sala Torra
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Megan Othus
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; SWOG Statistical Center, Seattle, Washington
| | | | - Brent Wood
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Ilan Kirsch
- Adaptive Biotechnologies, Seattle, Washington
| | - Harlan Robins
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Lan Beppu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Margaret R O'Donnell
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Stephen J Forman
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Frederick R Appelbaum
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jerald P Radich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
39
|
Tembhare PR, Ghogale S, Ghatwai N, Badrinath Y, Kunder N, Patkar NV, Bibi AR, Chatterjee G, Arora B, Narula G, Banawali S, Deshpande N, Amare P, Gujral S, Subramanian PG. Evaluation of new markers for minimal residual disease monitoring in B-cell precursor acute lymphoblastic leukemia: CD73 and CD86 are the most relevant new markers to increase the efficacy of MRD 2016; 00B: 000-000. CYTOMETRY PART B-CLINICAL CYTOMETRY 2016; 94:100-111. [PMID: 27718302 DOI: 10.1002/cyto.b.21486] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 10/02/2016] [Accepted: 10/05/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Multiparametric flow cytometry (MFC) is a popular technique for minimal residual disease (MRD) analysis. However, its applicability is still limited to 90% of B-cell precursor acute lymphoblastic leukemia (BCPALL) due to two major issues, i.e. a proportion of cases do not express adequate leukemia associated immunophenotype (LAIPs) with currently used markers and drug-induced antigen modulation. Hence, the incorporation of additional reliable markers is required for the further improvement of MFC-based MRD evaluation. We studied the utility of new markers in improvising MFC-based MRD detection in BCPALL. METHODS Expression-patterns of six new markers, i.e. CD24, CD44, CD72, CD73, CD86, and CD200 were studied in leukemic-blasts from ninety childhood BCPALL patients and in hematogones from 20 uninvolved staging bone marrow (BM) and ten postinduction non-BCPALL BM samples using eight-color MFC. The utility of these new markers in the day 35 postinduction MRD evaluation was determined. RESULTS Frequencies of LAIPs of CD73, CD86, CD72, CD44, CD200, and CD24 in diagnostic samples were 76.7, 56.7, 55.6, 50, 28.9, and 20%, respectively. Differential expression of all new markers was highly significant (P < 0.01) between early (CD10+ CD19+ CD34+) hematogones, late (CD10+ CD19+ CD34-) hematogones and BCPALL blasts except between early hematogones and BCPALL blasts for CD200 (P = 0.1). In MRD-positive samples, CD73 showed the maximum (83%) frequency of LAIP and CD86 showed the highest (100%) stability of aberrant expression. Inclusion of CD73 and CD86 increased the applicability of MFC-MRD assay to 98.9% MRD samples. CONCLUSION CD73 and CD86 are the most relevant markers to incorporate in the routine MRD evaluation of BCPALL. © 2016 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Prashant R Tembhare
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Sitaram Ghogale
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Nisha Ghatwai
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Yajamanam Badrinath
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Nikesh Kunder
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Nikhil V Patkar
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Asma R Bibi
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Gaurav Chatterjee
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Brijesh Arora
- Department of Pediatric Oncology, Tata Memorial Center, Main Building, Ground floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Gaurav Narula
- Department of Pediatric Oncology, Tata Memorial Center, Main Building, Ground floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Shripad Banawali
- Department of Pediatric Oncology, Tata Memorial Center, Main Building, Ground floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Nilesh Deshpande
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Prathibha Amare
- Department of Cancer Cytogenetics, Tata Memorial Center, Mumbaim, Room 726, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Sumeet Gujral
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| | - Papagudi G Subramanian
- Hematopathology Laboratory, Tata Memorial Center, Mumbai, Room 727, Hematopathology Laboratory, Annexe Building, 7th Floor, Tata Memorial Hospital, Parel, 400012, Mumbai, India
| |
Collapse
|
40
|
Abstract
The aim of this study was to identify key markers of minimal residual disease (MRD) in childhood Acute Lymphoblastic Leukemia (ALL). Bone marrow samples were collected at presentation from 139 patients with newly diagnosed B-lineage ALL. On the basis of the expression of CD19, CD10, and CD34 antigens by bone marrow cells, combined with the terminal deoxynucleotide transferase (TdT), CD38, CD45, CD58, CD21, CD66c, CD22, and CD33 expression patterns characterized at diagnosis, leukemia-associated immunophenotypes (LAIPs) were identified. One hundred thirty-nine patients with a median age of 4.3 years were screened with 4-color flow cytometry MRD screening, and 119 of them exhibited 1 or more LAIP suitable for further monitoring, constituting a coverage rate of 85.6%. Only 20 of the 139 (14.4%) had no LAIP identified for follow-up. The most applicable antibody combination was TdT/CD10/CD34/CD19 (87/139, 62.6%), followed by CD38/CD10/CD34/CD19 (85/139, 61.2%) and CD45/CD10/CD34/CD19 (58/139, 41.7%). We have identified a relatively effective MRD panel, combined with TdT, CD38, and CD45 as key markers, that is applicable to the majority of newly diagnosed B-lineage ALL.
Collapse
|
41
|
Bommannan K, Totadri S, Sachdeva MUS, Naseem S, Trehan A, Varma N. p210 BCR-ABL1 positive pediatric B-lineage acute lymphoblastic leukemia presenting with hypercalcemia. Leuk Lymphoma 2016; 58:501-502. [PMID: 27339816 DOI: 10.1080/10428194.2016.1196814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Karthik Bommannan
- a Department of Hematology , PostGraduate Institute of Medical Education and Research (PGIMER) , Chandigarh , India
| | - Sidharth Totadri
- b Pediatric Hematology-Oncology Unit , PostGraduate Institute of Medical Education and Research (PGIMER) , Chandigarh , India
| | - Man Updesh Singh Sachdeva
- a Department of Hematology , PostGraduate Institute of Medical Education and Research (PGIMER) , Chandigarh , India
| | - Shano Naseem
- a Department of Hematology , PostGraduate Institute of Medical Education and Research (PGIMER) , Chandigarh , India
| | - Amita Trehan
- b Pediatric Hematology-Oncology Unit , PostGraduate Institute of Medical Education and Research (PGIMER) , Chandigarh , India
| | - Neelam Varma
- a Department of Hematology , PostGraduate Institute of Medical Education and Research (PGIMER) , Chandigarh , India
| |
Collapse
|
42
|
Bartram J, Wade R, Vora A, Hancock J, Mitchell C, Kinsey S, Steward C, Moppett J, Goulden N. Excellent outcome of minimal residual disease-defined low-risk patients is sustained with more than 10 years follow-up: results of UK paediatric acute lymphoblastic leukaemia trials 1997-2003. Arch Dis Child 2016; 101:449-54. [PMID: 26865705 DOI: 10.1136/archdischild-2015-309617] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/20/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Minimal residual disease (MRD) is defined as the presence of sub-microscopic levels of leukaemia. Measurement of MRD from bone marrow at the end of induction chemotherapy (day 28) for childhood acute lymphoblastic leukaemia (ALL) can highlight a large group of patients (>40%) with an excellent (>90%) short-term event-free survival (EFS). However, follow-up in recent published trials is relatively short, raising concerns about using this result to infer the safety of further therapy reduction in the future. METHODS We examined MRD data on 225 patients treated on one of three UKALL trials between 1997 and 2003 to assess the long-term (>10 years follow-up) outcome of those patients who had low-risk MRD (<0.01%) at day 28. RESULTS Our pilot data define a cohort of 53% of children with MRD <0.01% at day 28 who have an EFS of 91% and long-term overall survival of 97%. Of 120 patients with day-28 MRD <0.01% and extended follow-up, there was one death due to treatment-related toxicity, one infectious death while in complete remission, and four relapse deaths. CONCLUSIONS The excellent outcome for childhood ALL in patients with MRD <0.01% after induction chemotherapy is sustained for more than 10 years from diagnosis. This supports the potential exploration of further reduction of therapy in this group, in an attempt to reduce treatment-related mortality and late effects.
Collapse
Affiliation(s)
- Jack Bartram
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Rachel Wade
- Clinical Trial Service Unit, University of Oxford, Oxford, UK
| | - Ajay Vora
- Department of Haematology, Sheffield Children's Hospital, Sheffield, UK
| | - Jeremy Hancock
- Bristol Genetics Laboratory, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Chris Mitchell
- Paediatric Haematology and Oncology, John Radcliffe Hospital, Oxford, UK
| | - Sally Kinsey
- Department of Paediatric Haematology, St James' University Hospital, Leeds, UK
| | - Colin Steward
- Department of Paediatric Haematology/Oncology, Royal Hospital for Children, Bristol, UK
| | - John Moppett
- Department of Paediatric Haematology/Oncology, Royal Hospital for Children, Bristol, UK
| | - Nick Goulden
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| |
Collapse
|
43
|
Current Strategies for the Detection of Minimal Residual Disease in Childhood Acute Lymphoblastic Leukemia. Mediterr J Hematol Infect Dis 2016; 8:e2016024. [PMID: 27158437 PMCID: PMC4848021 DOI: 10.4084/mjhid.2016.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/25/2016] [Indexed: 01/09/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in children. Current treatment strategies for childhood ALL result in long-term remission for approximately 90% of patients. However, the therapeutic response is worse among those who relapse. Several risk stratification approaches based on clinical and biological aspects have been proposed to intensify treatment in patients with high risk of relapse and reduce toxicity on those with a greater probability of cure. The detection of residual leukemic cells (minimal residual disease, MRD) is the most important prognostic factor to identify high-risk patients, allowing redefinition of chemotherapy. In the last decades, several standardized research protocols evaluated MRD using immunophenotyping by flow cytometry and/or real-time quantitative polymerase chain reaction at different time points during treatment. Both methods are highly sensitive (10−3 a 10−5), but expensive, complex, and, because of that, require qualified staff and frequently are restricted to reference centers. The aim of this article was to review technical aspects of immunophenotyping by flow cytometry and real-time quantitative polymerase chain reaction to evaluate MRD in ALL.
Collapse
|
44
|
Sever C, Abbott CL, de Baca ME, Khoury JD, Perkins SL, Reichard KK, Taylor A, Terebelo HR, Colasacco C, Rumble RB, Thomas NE. Bone Marrow Synoptic Reporting for Hematologic Neoplasms: Guideline From the College of American Pathologists Pathology and Laboratory Quality Center. Arch Pathol Lab Med 2016; 140:932-49. [PMID: 26905483 DOI: 10.5858/arpa.2015-0450-sa] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT -There is ample evidence from the solid tumor literature that synoptic reporting improves accuracy and completeness of relevant data. No evidence-based guidelines currently exist for synoptic reporting for bone marrow samples. OBJECTIVE -To develop evidence-based recommendations to standardize the basic components of a synoptic report template for bone marrow samples. DESIGN -The College of American Pathologists Pathology and Laboratory Quality Center convened a panel of experts in hematopathology to develop recommendations. A systematic evidence review was conducted to address 5 key questions. Recommendations were derived from strength of evidence, open comment feedback, and expert panel consensus. RESULTS -Nine guideline statements were established to provide pathology laboratories with a framework by which to develop synoptic reporting templates for bone marrow samples. The guideline calls for specific data groups in the synoptic section of the pathology report; provides a list of evidence-based parameters for key, pertinent elements; and addresses ancillary testing. CONCLUSION -A framework for bone marrow synoptic reporting will improve completeness of the final report in a manner that is clear, succinct, and consistent among institutions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Nicole E Thomas
- From the Department of Hematopathology, Pathology Associates of Albuquerque, Albuquerque, New Mexico (Dr Sever); the Department of Pathology, Berkshire Medical Center, Pittsfield, Massachusetts (Dr Abbott); Medical Laboratory Associates, Seattle, Washington (Dr de Baca); the Department of Pathology, University of Texas MD Anderson Cancer Center, Houston (Dr Khoury); the Department of Pathology, University of Utah, Salt Lake City (Dr Perkins); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Reichard); Utah Pathology Services, Inc, Salt Lake City (Dr Taylor); the Department of Hematology/Medical Oncology, Newland Medical Associates, Novi, Michigan (Dr Terebelo); the Departments of Governance (Ms Colasacco) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; and the Quality and Guidelines Department, American Society of Clinical Oncology, Alexandria, Virginia (Mr Rumble)
| |
Collapse
|
45
|
Irving JAE. Towards an understanding of the biology and targeted treatment of paediatric relapsed acute lymphoblastic leukaemia. Br J Haematol 2015; 172:655-66. [PMID: 26568036 DOI: 10.1111/bjh.13852] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Acute lymphoblastic leukaemia is the most common childhood cancer and for those children who relapse, prognosis is poor and new therapeutic strategies are needed. Recurrent pathways implicated in relapse include RAS, JAK STAT, cell cycle, epigenetic regulation, B cell development, glucocorticoid response, nucleotide metabolism and DNA repair. Targeting these pathways is a rational therapeutic strategy and may deliver novel, targeted therapies into the clinic. Relapse often stems from a minor clone present at diagnosis and thus analysis of persisting leukaemia during upfront therapy may allow targeted drug intervention to prevent relapse.
Collapse
Affiliation(s)
- Julie A E Irving
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| |
Collapse
|
46
|
Ikoma MRV, Beltrame MP, Ferreira SIACP, Souto EX, Malvezzi M, Yamamoto M. Proposal for the standardization of flow cytometry protocols to detect minimal residual disease in acute lymphoblastic leukemia. Rev Bras Hematol Hemoter 2015; 37:406-13. [PMID: 26670404 PMCID: PMC4678914 DOI: 10.1016/j.bjhh.2015.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 07/23/2015] [Accepted: 07/27/2015] [Indexed: 01/26/2023] Open
Abstract
Minimal residual disease is the most powerful predictor of outcome in acute leukemia and is useful in therapeutic stratification for acute lymphoblastic leukemia protocols. Nowadays, the most reliable methods for studying minimal residual disease in acute lymphoblastic leukemia are multiparametric flow cytometry and polymerase chain reaction. Both provide similar results at a minimal residual disease level of 0.01% of normal cells, that is, detection of one leukemic cell in up to 10,000 normal nucleated cells. Currently, therapeutic protocols establish the minimal residual disease threshold value at the most informative time points according to the appropriate methodology employed. The expertise of the laboratory in a cancer center or a cooperative group could be the most important factor in determining which method should be used. In Brazil, multiparametric flow cytometry laboratories are available in most leukemia treatment centers, but multiparametric flow cytometry processes must be standardized for minimal residual disease investigations in order to offer reliable and reproducible results that ensure quality in the clinical application of the method. The Minimal Residual Disease Working Group of the Brazilian Society of Bone Marrow Transplantation (SBTMO) was created with that aim. This paper presents recommendations for the detection of minimal residual disease in acute lymphoblastic leukemia based on the literature and expertise of the laboratories who participated in this consensus, including pre-analytical and analytical methods. This paper also recommends that both multiparametric flow cytometry and polymerase chain reaction are complementary methods, and so more laboratories with expertise in immunoglobulin/T cell receptor (Ig/TCR) gene assays are necessary in Brazil.
Collapse
Affiliation(s)
| | | | | | | | | | - Mihoko Yamamoto
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | |
Collapse
|
47
|
DiGiuseppe JA, Tadmor MD, Pe’er D. Detection of minimal residual disease in B lymphoblastic leukemia using viSNE. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2015; 88:294-304. [PMID: 25974871 PMCID: PMC5981136 DOI: 10.1002/cyto.b.21252] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/03/2015] [Accepted: 05/11/2015] [Indexed: 11/05/2022]
Abstract
BACKGROUND Minimal residual disease (MRD) following treatment is a robust prognostic marker in B lymphoblastic leukemia. However, the detection of MRD by flow cytometric immunophenotyping is technically challenging, and an automated method to detect MRD is therefore desirable. viSNE, a recently developed computational tool based on the t-Distributed Stochastic Neighbor Embedding (t-SNE) algorithm, has been shown to be capable of detecting synthetic "MRD-like" populations of leukemic cells created in vitro, but whether viSNE can facilitate the immunophenotypic detection of MRD in clinical samples has not been evaluated. METHODS We applied viSNE retrospectively to 8-color flow cytometric immunophenotyping data from normal bone marrow samples, and samples from B lymphoblastic leukemia patients with or without suspected MRD on the basis of conventional manual gating. RESULTS In each of 14 bone marrow specimens containing MRD or suspected MRD, viSNE identified a putative MRD population; an abnormal composite immunophenotype was confirmed for the putative MRD in each case. MRD populations were not identified by viSNE in control bone marrow samples from patients with increased normal B-cell precursors, or in post-treatment samples from B lymphoblastic leukemia patients who did not have detectable MRD by manual gating. CONCLUSION viSNE shows promise as an automated method to facilitate immunophenotypic MRD detection in patients treated for B lymphoblastic leukemia.
Collapse
Affiliation(s)
- Joseph A. DiGiuseppe
- Department of Pathology & Laboratory Medicine, Hartford Hospital, Hartford, Connecticut
| | - Michelle D. Tadmor
- Department of Biological Sciences, Columbia University, New York, New York
- Department of Systems Biology, Columbia University, New York, New York
| | - Dana Pe’er
- Department of Biological Sciences, Columbia University, New York, New York
- Department of Systems Biology, Columbia University, New York, New York
| |
Collapse
|
48
|
Karawajew L, Dworzak M, Ratei R, Rhein P, Gaipa G, Buldini B, Basso G, Hrusak O, Ludwig WD, Henze G, Seeger K, von Stackelberg A, Mejstrikova E, Eckert C. Minimal residual disease analysis by eight-color flow cytometry in relapsed childhood acute lymphoblastic leukemia. Haematologica 2015; 100:935-44. [PMID: 26001791 DOI: 10.3324/haematol.2014.116707] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 04/30/2015] [Indexed: 01/17/2023] Open
Abstract
Multiparametric flow cytometry is an alternative approach to the polymerase chain reaction method for evaluating minimal residual disease in treatment protocols for primary acute lymphoblastic leukemia. Given considerable differences between primary and relapsed acute lymphoblastic leukemia treatment regimens, flow cytometric assessment of minimal residual disease in relapsed leukemia requires an independent comprehensive investigation. In the present study we addressed evaluation of minimal residual disease by flow cytometry in the clinical trial for childhood relapsed acute lymphoblastic leukemia using eight-color flow cytometry. The major challenge of the study was to reliably identify low amounts of residual leukemic cells against the complex background of regeneration, characteristic of follow-up samples during relapse treatment. In a prospective study of 263 follow-up bone marrow samples from 122 patients with B-cell precursor acute lymphoblastic leukemia, we tested various B-cell markers, adapted the antibody panel to the treatment protocol, and evaluated its performance by a blinded parallel comparison with the polymerase chain reaction data. The resulting eight-color single-tube panel showed a consistently high overall concordance (P<0.001) and, under optimal conditions, sensitivity similar to that of the reference polymerase chain reaction method. Overall, evaluation of minimal residual disease by flow cytometry can be successfully integrated into the clinical management of relapsed childhood acute lymphoblastic leukemia either as complementary to the polymerase chain reaction or as an independent risk stratification tool. ALL-REZ BFM 2002 clinical trial information: NCT00114348.
Collapse
Affiliation(s)
- Leonid Karawajew
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin, Berlin, Germany
| | - Michael Dworzak
- St. Anna Children's Hospital and Children's Cancer Research Institute, Department of Pediatrics, Medical University of Vienna, Austria
| | - Richard Ratei
- Robert-Roessle-Clinic in the HELIOS Klinikum Berlin, Germany
| | - Peter Rhein
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin, Berlin, Germany
| | - Giuseppe Gaipa
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca, Ospedale San Gerardo, Monza, Italy
| | - Barbara Buldini
- Laboratory of Pediatric Onco-Hematology, Department of Pediatrics, University Hospital of Padova, Italy
| | - Giuseppe Basso
- Laboratory of Pediatric Onco-Hematology, Department of Pediatrics, University Hospital of Padova, Italy
| | - Ondrej Hrusak
- Department of Pediatric Hematology and Oncology, Charles University 2 Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | | | - Günter Henze
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin, Berlin, Germany
| | - Karl Seeger
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin, Berlin, Germany
| | - Arend von Stackelberg
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin, Berlin, Germany
| | - Ester Mejstrikova
- Department of Pediatric Hematology and Oncology, Charles University 2 Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Cornelia Eckert
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
49
|
Salari F, Shahjahani M, Shahrabi S, Saki N. Minimal residual disease in acute lymphoblastic leukemia: optimal methods and clinical relevance, pitfalls and recent approaches. Med Oncol 2014; 31:266. [PMID: 25287907 DOI: 10.1007/s12032-014-0266-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 09/20/2014] [Indexed: 11/29/2022]
Abstract
After advances in experimental and clinical testing, minimal residual disease (MRD) assay results are considered a determining factor in treatment of acute lymphoblastic leukemia patients. According to MRD assay results, bone marrow (BM) leukemic burden and the rate of its decline after treatment can be directly evaluated. Detailed knowledge of the leukemic burden in BM can minimize toxicity and treatment complications in patients by tailoring the therapeutic dose based on patients' conditions. In addition, reduction of MRD before allo-HSCT is an important prerequisite for reception of transplant by the patient. In direct examination of MRD by morphological methods (even by a professional hematologist), leukemic cells can be under- or over-estimated due to similarity with hematopoietic precursor cells. As a result, considering the importance of MRD, it is necessary to use other methods including flow cytometry, polymerase chain reaction (PCR) amplification and RQ-PCR to detect MRD. Each of these methods has its own advantages and disadvantages in terms of accuracy and sensitivity. In this review article, different MRD assay methods and their sensitivity, correlation of MRD assay results with clinical symptoms of the patient as well as pitfalls in results of these methods are evaluated. In the final section, recent advances in MRD have been addressed.
Collapse
Affiliation(s)
- Fatemeh Salari
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | | | | |
Collapse
|
50
|
Johansson U, Bloxham D, Couzens S, Jesson J, Morilla R, Erber W, Macey M. Guidelines on the use of multicolour flow cytometry in the diagnosis of haematological neoplasms. British Committee for Standards in Haematology. Br J Haematol 2014; 165:455-88. [PMID: 24620735 DOI: 10.1111/bjh.12789] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|