1
|
van der Velden VHJ, Dombrink I, Alten J, Cazzaniga G, Clappier E, Drandi D, Eckert C, Fronkova E, Hancock J, Kotrova M, Kraemer R, Montonen M, Pfeifer H, Pott C, Raff T, Trautmann H, Cavé H, Schäfer BW, van Dongen JJM, Trka J, Brüggemann M. Analysis of measurable residual disease by IG/TR gene rearrangements: quality assurance and updated EuroMRD guidelines. Leukemia 2024; 38:1315-1322. [PMID: 38744919 PMCID: PMC11147754 DOI: 10.1038/s41375-024-02272-0] [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: 01/13/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
Minimal/measurable residual disease (MRD) diagnostics using real-time quantitative PCR analysis of rearranged immunoglobulin and T-cell receptor gene rearrangements are nowadays implemented in most treatment protocols for patients with acute lymphoblastic leukemia (ALL). Within the EuroMRD Consortium, we aim to provide comparable, high-quality MRD diagnostics, allowing appropriate risk-group classification for patients and inter-protocol comparisons. To this end, we set up a quality assessment scheme, that was gradually optimized and updated over the last 20 years, and that now includes participants from around 70 laboratories worldwide. We here describe the design and analysis of our quality assessment scheme. In addition, we here report revised data interpretation guidelines, based on our newly generated data and extensive discussions between experts. The main novelty is the partial re-definition of the "positive below quantitative range" category by two new categories, "MRD low positive, below quantitative range" and "MRD of uncertain significance". The quality assessment program and revised guidelines will ensure reproducible and accurate MRD data for ALL patients. Within the Consortium, similar programs and guidelines have been introduced for other lymphoid diseases (e.g., B-cell lymphoma), for new technological platforms (e.g., digital droplet PCR or Next-Generation Sequencing), and for other patient-specific MRD PCR-based targets (e.g., fusion genes).
Collapse
Affiliation(s)
- Vincent H J van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Isabel Dombrink
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Julia Alten
- Department of Pediatrics, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Giovanni Cazzaniga
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine, University of Milano-Bicocca, Monza, Italy
| | - Emmanuelle Clappier
- Hematology Laboratory, Saint-Louis Hospital, Paris Cité University, Paris, France
- Université Paris-Cité, Paris, France
| | - Daniela Drandi
- Department of Molecular Biotechnology and health sciences, Hematology Division, University of Torino, Torino, Italy
| | - Cornelia Eckert
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eva Fronkova
- CLIP, Department of Pediatric Hematology and Oncology, Second Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jeremy Hancock
- Bristol MRD Group, Bristol Genetics Laboratory, Southmead Hospital, Bristol, UK
| | - Michaela Kotrova
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Rebekka Kraemer
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Mirkka Montonen
- Tyks Laboratories, Genomics Department, Turku University Hospital, Turku, Finland
| | - Heike Pfeifer
- Department of Hematology, University Hospital Frankfurt, Frankfurt, Germany
| | - Christiane Pott
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thorsten Raff
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
- Military Medical City Hospital, Doha, Qatar
| | - Heiko Trautmann
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Hélène Cavé
- Hematology Laboratory, Saint-Louis Hospital, Paris Cité University, Paris, France
- Department of Genetics, University Hospital Robert Debré, Paris, France
| | | | - Jacques J M van Dongen
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC, USAL-CSIC-FICUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
- European Scientific foundation for Laboratory Hemato Oncology (ESLHO), Zutphen, The Netherlands
- Department of Immunology, LUMC, Leiden, The Netherlands
| | - Jan Trka
- CLIP, Department of Pediatric Hematology and Oncology, Second Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Monika Brüggemann
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
2
|
Chen J, Gale RP, Hu Y, Yan W, Wang T, Zhang W. Measurable residual disease (MRD)-testing in haematological and solid cancers. Leukemia 2024; 38:1202-1212. [PMID: 38637690 PMCID: PMC11147778 DOI: 10.1038/s41375-024-02252-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Affiliation(s)
- Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wen Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tiantian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
3
|
Campbell M, Kiss C, Zimmermann M, Riccheri C, Kowalczyk J, Felice MS, Kuzmanovic M, Kovacs G, Kosmidis H, Gonzalez A, Bilic E, Castillo L, Kolenova A, Jazbec J, Popa A, Konstantinov D, Kappelmayer J, Szczepanski T, Dworzak M, Buldini B, Gaipa G, Marinov N, Rossi J, Nagy A, Gaspar I, Stary J, Schrappe M. Childhood Acute Lymphoblastic Leukemia: Results of the Randomized Acute Lymphoblastic Leukemia Intercontinental-Berlin-Frankfurt-Münster 2009 Trial. J Clin Oncol 2023:JCO2201760. [PMID: 37141547 DOI: 10.1200/jco.22.01760] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
PURPOSE The International Berlin-Frankfurt-Münster (BFM) study group conducted a study on pediatric acute lymphoblastic leukemia (ALL). Minimal residual disease (MRD) was assessed using flow cytometry (FCM), and the impact of early intensification and methotrexate (MTX) dose on survival was evaluated. PATIENTS AND METHODS We included 6,187 patients younger than 19 years. MRD by FCM refined the risk group definition previously used in the ALL intercontinental-BFM 2002 study on the basis of age, WBC count, unfavorable genetic aberrations, and treatment response measured morphologically. Patients at intermediate risk (IR) and high risk (HR) were randomly assigned to protocol augmented protocol I phase B (IB) versus IB regimen. MTX doses of 2 versus 5 g/m2 every 2 weeks, four times, were evaluated in precursor B-cell-ALL (pcB-ALL) IR. RESULTS The 5-year event-free survival (EFS ± SE) and overall survival (OS ± SE) rates were 75.2% ± 0.6% and 82.6% ± 0.5%, respectively. Their values in risk groups were standard risk (n = 624), 90.7% ± 1.4% and 94.7% ± 1.1%; IR (n = 4,111), 77.9% ± 0.7% and 85.7% ± 0.6%; and HR (n = 1,452), 60.8% ± 1.5% and 68.4% ± 1.4%, respectively. MRD by FCM was available in 82.6% of cases. The 5-year EFS rates in patients randomly assigned to protocol IB (n = 1,669) and augmented IB (n = 1,620) were 73.6% ± 1.2% and 72.8% ± 1.2%, respectively (P = .55), while those in patients receiving MTX doses of 2 g/m2 (n = 1,056) and MTX 5 g/m2 (n = 1,027) were 78.8% ± 1.4% and 78.9% ± 1.4%, respectively (P = .84). CONCLUSION The MRDs were successfully assessed using FCM. An MTX dose of 2 g/m2 was effective in preventing relapse in non-HR pcB-ALL. Augmented IB showed no advantages over the standard IB.
Collapse
Affiliation(s)
- Myriam Campbell
- Department of Pediatric Hematology and Oncology, Hospital Roberto del Rio, Universidad de Chile, Chilean National Pediatric Oncology Group, PINDA, Santiago, Chile
| | - Csongor Kiss
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Martin Zimmermann
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Cecilia Riccheri
- Argentine Group for the Treatment of Acute Leukemia, GATLA, Buenos Aires, Argentina
| | - Jerzy Kowalczyk
- Department of Pediatric, Hematology, Oncology, and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Maria S Felice
- Hematology and Oncology Department, Hospital de Pediatría Prof. Dr Juan P. Garrahan, SAHOP, Buenos Aires, Argentina
| | - Milos Kuzmanovic
- Mother and Child Health Care Institute of Serbia "Dr Vukan Cupic", Faculty of Medicine, Belgrade, Serbia
| | - Gabor Kovacs
- 2nd Department of Pediatrics Semmelweis University, Budapest, Hungary
| | - Helen Kosmidis
- Pediatric and Adolescent Oncology Clinic, Children's Hospital MITERA, Athens, Greece
| | | | - Ernest Bilic
- School of Medicine Division of Pediatric Hematology and Oncology, University Hospital Center, University of Zagreb, Zagreb, Croatia
| | - Luis Castillo
- Pediatric Hemato-Oncology Department, Hospital Pereira Rossell, Pérez Scremini Foundation, Montevideo, Uruguay
| | - Alexandra Kolenova
- Department of Pediatric Hematology and Oncology, National Institute of Children's Diseases and Medical School, Comenius University, Bratislava, Slovakia
| | - Janez Jazbec
- University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Alexander Popa
- Pediatric Oncology and Hematology Research Institute of N.N.Blokhin National Cancer Research Center, Center, Moscow, Russia
| | - Dobrin Konstantinov
- Pediatric Hematology & Oncology Department, University Hospital "Tsaritsa Johanna-ISUL", Sofia, Bulgaria
| | - Janos Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia, Katowice, Poland
| | - Michael Dworzak
- St Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Barbara Buldini
- Mother and Child's Health Department, Division of Pediatric Hematology, Oncology and Stem Cell Transplant, University of Padova, Padova, Veneto, Italy
| | - Giuseppe Gaipa
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Neda Marinov
- Chilean National Pediatric Oncology Group, PINDA, Hospital Roberto del Rio/Universidad de Chile, Santiago, Chile
- Hospital del Salvador, Universidad de Chile, Santiago, Chile
| | - Jorge Rossi
- Immunology and Rheumatology Department, Hospital de Pediatría Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina
| | - Attila Nagy
- Department of Interventional Epidemiology, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Imre Gaspar
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jan Stary
- Department of Pediatric Hematology and Oncology Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Martin Schrappe
- Department of Pediatric and Adolescent Medicine, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
4
|
Brum da Silva Nunes V, Kehl Dias C, Nathali Scholl J, Nedel Sant'Ana A, de Fraga Dias A, Granero Farias M, Alegretti AP, Sosnoski M, Esteves Daudt L, Bohns Michalowski M, Oliveira Battastini AM, Paz AA, Figueiró F. Lymphocytes from B-acute lymphoblastic leukemia patients present differential regulation of the adenosinergic axis depending on risk stratification. Discov Oncol 2022; 13:143. [PMID: 36581667 PMCID: PMC9800668 DOI: 10.1007/s12672-022-00602-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Although risk-stratified chemotherapy regimens improve B-cell acute lymphoblastic leukemia (B-ALL) clinical outcome, relapse occurs in a significant number of cases. The identification of new therapeutic targets as well as prognostic and diagnostic biomarkers can improve B-ALL patients' clinical outcomes. Purinergic signaling is an important pathway in cancer progression, however the expression of ectonucleotidases and their impact on immune cells in B-ALL lacks exploration. We aimed to analyze the expression of ectonucleotidases in B-ALL patients' lymphocyte subpopulations. METHODS Peripheral blood samples from 15 patients diagnosed with B-ALL were analyzed. Flow cytometry was used to analyze cellularity, expression level of CD38, CD39, and CD73, and frequency of [Formula: see text], and [Formula: see text] in lymphocyte subpopulations. Plasma was used for cytokines (by CBA kit) and adenine nucleosides/nucleotides detection (by HPLC). RESULTS Comparing B-ALL patients to health donors, we observed an increase of CD4 + and CD8 + T-cells. In addition, a decrease in CD38 expression in B and Treg subpopulations and an increase in CD39+ CD73+ frequency in Breg and CD8+ T-cells. Analyzing cytokines and adenine nucleosides/nucleotides, we found a decrease in TNF, IL-1β, and ADO concentrations, together with an increase in AMP in B-ALL patients' plasma. CONCLUSION As immunomodulators, the expression of ectonucleotidases might be associated with activation states, as well as the abundance of different cellular subsets. We observed a pro-tumor immunity expression profile in B-ALL patients at diagnosis, being associated with cell exhaustion and immune evasion in B-ALL.
Collapse
Affiliation(s)
- Vitória Brum da Silva Nunes
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Camila Kehl Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Juliete Nathali Scholl
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Alexia Nedel Sant'Ana
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Amanda de Fraga Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | - Ana Paula Alegretti
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Monalisa Sosnoski
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Liane Esteves Daudt
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Mariana Bohns Michalowski
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, UFRGS, Porto Alegre, RS, 90035-003, Brazil
| | - Ana Maria Oliveira Battastini
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | - Fabrício Figueiró
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil.
| |
Collapse
|
5
|
Immune Gene Rearrangements: Unique Signatures for Tracing Physiological Lymphocytes and Leukemic Cells. Genes (Basel) 2021; 12:genes12070979. [PMID: 34198966 PMCID: PMC8329920 DOI: 10.3390/genes12070979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023] Open
Abstract
The tremendous diversity of the human immune repertoire, fundamental for the defense against highly heterogeneous pathogens, is based on the ingenious mechanism of immune gene rearrangements. Rearranged immune genes encoding the immunoglobulins and T-cell receptors and thus determining each lymphocyte's antigen specificity are very valuable molecular markers for tracing malignant or physiological lymphocytes. One of their most significant applications is tracking residual leukemic cells in patients with lymphoid malignancies. This so called 'minimal residual disease' (MRD) has been shown to be the most important prognostic factor across various leukemia subtypes and has therefore been given enormous attention. Despite the current rapid development of the molecular methods, the classical real-time PCR based approach is still being regarded as the standard method for molecular MRD detection due to the cumbersome standardization of the novel approaches currently in progress within the EuroMRD and EuroClonality NGS Consortia. Each of the molecular methods, however, poses certain benefits and it is therefore expectable that none of the methods for MRD detection will clearly prevail over the others in the near future.
Collapse
|
6
|
Dander E, Fallati A, Gulić T, Pagni F, Gaspari S, Silvestri D, Cricrì G, Bedini G, Portale F, Buracchi C, Starace R, Pasqualini F, D'Angiò M, Brizzolara L, Maglia O, Mantovani A, Garlanda C, Valsecchi MG, Locatelli F, Biondi A, Bottazzi B, Allavena P, D'Amico G. Monocyte-macrophage polarization and recruitment pathways in the tumour microenvironment of B-cell acute lymphoblastic leukaemia. Br J Haematol 2021; 193:1157-1171. [PMID: 33713428 DOI: 10.1111/bjh.17330] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
B-cell acute lymphoblastic leukaemia (B-ALL) reprograms the surrounding bone marrow (BM) stroma to create a leukaemia-supportive niche. To elucidate the contribution of immune cells to the leukaemic microenvironment, we investigated the involvement of monocyte/macrophage compartments, as well as several recruitment pathways in B-ALL development. Immunohistochemistry analyses showed that CD68-expressing macrophages were increased in leukaemic BM biopsies, compared to controls and predominantly expressed the M2-like markers CD163 and CD206. Furthermore, the "non-classical" CD14+ CD16++ monocyte subset, expressing high CX3CR1 levels, was significantly increased in B-ALL patients' peripheral blood. CX3CL1 was shown to be significantly upregulated in leukaemic BM plasma, thus providing an altered migratory pathway possibly guiding NC monocyte recruitment into the BM. Additionally, the monocyte/macrophage chemoattractant chemokine ligand 2 (CCL2) strongly increased in leukaemic BM plasma, possibly because of the interaction of leukaemic cells with mesenchymal stromal cells and vascular cells and due to a stimulatory effect of leukaemia-related inflammatory mediators. C5a, a macrophage chemoattractant and M2-polarizing factor, further appeared to be upregulated in the leukaemic BM, possibly as an effect of PTX3 decrease, that could unleash complement cascade activation. Overall, deregulated monocyte/macrophage compartments are part of the extensive BM microenvironment remodelling at B-ALL diagnosis and could represent valuable targets for novel treatments to be coupled with classical chemotherapy.
Collapse
Affiliation(s)
- Erica Dander
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Alessandra Fallati
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Tamara Gulić
- IRCCS, Humanitas Clinical and Research Center, Rozzano (Mi), Italy
| | - Fabio Pagni
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Stefania Gaspari
- Department of Pediatric Hematology-Oncology, IRCCS Bambino Gesù Children's Hospital, Sapienza, University of Rome, Rome, Italy
| | - Daniela Silvestri
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Giulia Cricrì
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Gloria Bedini
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Federica Portale
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Chiara Buracchi
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Rita Starace
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Fabio Pasqualini
- IRCCS, Humanitas Clinical and Research Center, Rozzano (Mi), Italy
| | - Mariella D'Angiò
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Lisa Brizzolara
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Oscar Maglia
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Alberto Mantovani
- IRCCS, Humanitas Clinical and Research Center, Rozzano (Mi), Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele - Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Cecilia Garlanda
- IRCCS, Humanitas Clinical and Research Center, Rozzano (Mi), Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele - Milan, Italy
| | - Maria Grazia Valsecchi
- Center of Bioinformatics, Biostatistics and Bioimaging, School of Medicine and Surgery, University of Milano Bicocca, Monza, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology-Oncology, IRCCS Bambino Gesù Children's Hospital, Sapienza, University of Rome, Rome, Italy
| | - Andrea Biondi
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Barbara Bottazzi
- IRCCS, Humanitas Clinical and Research Center, Rozzano (Mi), Italy
| | - Paola Allavena
- IRCCS, Humanitas Clinical and Research Center, Rozzano (Mi), Italy
| | - Giovanna D'Amico
- Centro Ricerca Tettamanti, Pediatric Dep, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| |
Collapse
|
7
|
Alecsa MS, Moscalu M, Trandafir LM, Ivanov AV, Rusu C, Miron IC. Outcomes in Pediatric Acute Lymphoblastic Leukemia-A Single-Center Romanian Experience. J Clin Med 2020; 9:jcm9124052. [PMID: 33333966 PMCID: PMC7765371 DOI: 10.3390/jcm9124052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND This study evaluates the main (para)clinical aspects and outcomes in a group of Romanian children diagnosed with acute lymphoblastic leukemia (ALL), under the conditions of antileukemic treatment according to an adapted ALL IC Berlin-Frankfurt-Munster (BFM) 2002 protocol. METHODS We performed a retrospective single-center study of 125 children diagnosed with ALL between 2010 and 2016. Standard forms were used for data collection of variate clinical and paraclinical parameters. RESULTS The children were predominantly male (64.8%) and their median age at diagnosis was 5 years. A total of 107 patients were diagnosed with precursor B-cell acute lymphoblastic leukemia (BCP)-ALL and 18 with T-cell acute lymphoblastic leukemia T-ALL. Multiplex reverse transcription polymerase chain reaction RT-PCR assay for ETV6-RUNX1, BCR-ABL, E2A-PBX1, KMT2A-AFF1, and STIL-TAL1 fusion genes was performed in 111 patients. ETV6-RUNX1 translocation was detected in 18.9% of patients, while BCR-ABL1 and E2A-PBX1 rearrangements were seen in 2.7% and 3.6%, respectively. Complete remission at the end of induction phase was obtained in 89.6% of patients. The overall relapse rate was 11.2%, with 11 early and 3 late relapses. The 5-year overall survival rate in BCP-ALL was 81.6% and in T-ALL 71.4%. CONCLUSIONS The 5-year overall and event-free survival rates in our study were slightly lower than those reported in developed countries, so the patients' outcomes are encouraging.
Collapse
Affiliation(s)
- Mirabela-Smaranda Alecsa
- Department Mother and Child Care, Division of Neonatology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.-S.A.); (A.-V.I.); (C.R.); (I.-C.M.)
- Department of Pediatric Hematology and Oncology, Sf. Maria Children’s Emergency Hospital, 700309 Iasi, Romania
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, Division of Informatics and Medical Statistics, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (M.M.); (L.-M.T.)
| | - Laura-Mihaela Trandafir
- Department Mother and Child Care, Division of Neonatology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.-S.A.); (A.-V.I.); (C.R.); (I.-C.M.)
- Department of Pediatrics, Sf. Maria Children’s Emergency Hospital, 700309 Iasi, Romania
- Correspondence: (M.M.); (L.-M.T.)
| | - Anca-Viorica Ivanov
- Department Mother and Child Care, Division of Neonatology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.-S.A.); (A.-V.I.); (C.R.); (I.-C.M.)
- Department of Pediatrics, Sf. Maria Children’s Emergency Hospital, 700309 Iasi, Romania
| | - Cristina Rusu
- Department Mother and Child Care, Division of Neonatology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.-S.A.); (A.-V.I.); (C.R.); (I.-C.M.)
- Department of Medical Genetics, Sf. Maria Children’s Emergency Hospital, 700309 Iasi, Romania
| | - Ingrith-Crenguta Miron
- Department Mother and Child Care, Division of Neonatology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.-S.A.); (A.-V.I.); (C.R.); (I.-C.M.)
- Department of Pediatric Hematology and Oncology, Sf. Maria Children’s Emergency Hospital, 700309 Iasi, Romania
| |
Collapse
|
8
|
Lomov N, Zerkalenkova E, Lebedeva S, Viushkov V, Rubtsov MA. Cytogenetic and molecular genetic methods for chromosomal translocations detection with reference to the KMT2A/MLL gene. Crit Rev Clin Lab Sci 2020; 58:180-206. [PMID: 33205680 DOI: 10.1080/10408363.2020.1844135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acute leukemias (ALs) are often associated with chromosomal translocations, in particular, KMT2A/MLL gene rearrangements. Identification or confirmation of these translocations is carried out by a number of genetic and molecular methods, some of which are routinely used in clinical practice, while others are primarily used for research purposes. In the clinic, these methods serve to clarify diagnoses and monitor the course of disease and therapy. On the other hand, the identification of new translocations and the confirmation of known translocations are of key importance in the study of disease mechanisms and further molecular classification. There are multiple methods for the detection of rearrangements that differ in their principle of operation, the type of problem being solved, and the cost-result ratio. This review is intended to help researchers and clinicians studying AL and related chromosomal translocations to navigate this variety of methods. All methods considered in the review are grouped by their principle of action and include karyotyping, fluorescence in situ hybridization (FISH) with probes for whole chromosomes or individual loci, PCR and reverse transcription-based methods, and high-throughput sequencing. Another characteristic of the described methods is the type of problem being solved. This can be the discovery of new rearrangements, the determination of unknown partner genes participating in the rearrangement, or the confirmation of the proposed rearrangement between the two genes. We consider the specifics of the application, the basic principle of each method, and its pros and cons. To illustrate the application, examples of studying the rearrangements of the KMT2A/MLL gene, one of the genes that are often rearranged in AL, are mentioned.
Collapse
Affiliation(s)
- Nikolai Lomov
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Elena Zerkalenkova
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Svetlana Lebedeva
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Vladimir Viushkov
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail A Rubtsov
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Department of Biochemistry, Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
9
|
Jovanovska A, Martinova K, Kocheva S, Trajkova-Antevska Z, Coneska-Jovanova B, Panovska-Stavridis I, Stankovikj S, Trajkova S, Dimovski A. Clinical Significance of Minimal Residual Disease at the End of Remission Induction Therapy in Childhood Acute Lymphoblastic Leukemia. Open Access Maced J Med Sci 2019; 7:2818-2823. [PMID: 31844443 PMCID: PMC6901875 DOI: 10.3889/oamjms.2019.752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/04/2019] [Accepted: 08/05/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Detection of minimal residual disease (MRD) in the early phase of therapy is the most powerful predictor of relapse risk in children with acute lymphoblastic leukaemia (ALL). AIM We aimed to determine the significance of MRD at the end of remission induction therapy in the prediction of treatment outcome in children with ALL. METHODS Sixty-four consecutive patients aged 1-14 years with newly diagnosed ALL were enrolled in this study from January 2010 to October 2017. All patients were treated according to the ALL IC BFM 2002 protocol. MRD was detected at the end of remission induction therapy (day 33) by multiparameter 6-colour flow cytometry performed on bone marrow specimens with a sensitivity of 0.01%. RESULTS Overall, 42.2% of patients had detectable MRD on day 33 of therapy. MRD measurements were not significantly related to presenting characteristics but were associated with a poorer blast clearance on day 8 and 15 of remission induction therapy. Patients with negative MRD status on day 33 had a 5-year event-free survival of 94.6% compared with 76.1% for those with positive MRD status (P = 0.044). CONCLUSION MRD levels at the end of remission induction therapy measured by multiparameter flow cytometry have clinical significance in childhood ALL. High levels of MRD are strongly related to poor treatment outcome.
Collapse
Affiliation(s)
- Aleksandra Jovanovska
- Department of Hematology and Oncology, University Clinic for Children`s Diseases, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Kata Martinova
- Department of Hematology and Oncology, University Clinic for Children`s Diseases, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Svetlana Kocheva
- Department of Hematology and Oncology, University Clinic for Children`s Diseases, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Zorica Trajkova-Antevska
- Department of Hematology and Oncology, University Clinic for Children`s Diseases, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | | | - Irina Panovska-Stavridis
- University Clinic for Hematology, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Svetlana Stankovikj
- University Clinic for Hematology, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Sanja Trajkova
- University Clinic for Hematology, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Aleksandar Dimovski
- Faculty of Pharmacy, Ss Cyril and Methodius University of Skopje, Skopje, Skopje, Republic of Macedonia
| |
Collapse
|
10
|
Kamat G, Subramanian K, Apte S. Correlation of Day 8 Steroid Response with Bone Marrow Status Measured on Days 14 and 35, in Patients with Acute Lymphoblastic Leukemia Being Treated with BFM Protocol. Indian J Hematol Blood Transfus 2019; 35:635-639. [PMID: 31741614 DOI: 10.1007/s12288-019-01119-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 04/03/2019] [Indexed: 10/27/2022] Open
Abstract
Berlin-Frankfurt-Munster (BFM) protocol is commonly used in India for treatment of acute lymphoblastic leukemia (ALL). The present study was conducted to correlate day 8 steroid response with bone marrow status (by morphology and flow cytometry) at day 14 and day 35 of treatment. It was a prospective study which included all newly diagnosed ALL patients who visited hospital between March 2013 and February 2015 i.e. 2 years. On 8th day, the number of lymphoblasts was counted in the peripheral blood. Based on the number of blasts patients were classified as good steroid responders and poor steroid responders. Following pre-induction steroids patients were given induction therapy. During this phase on day 14 and day 35 bone marrow (BM) aspiration study was done. Later day 8 steroid response, Day 14 BM status and day 35 BM status were correlated. Results showed that there was a statistically significant correlation between day 8 steroid response and day 14 BM status (both by morphology and flow cytometry). There was no statistically significant correlation between day 8 steroid response and day 35 BM status (both by morphology and flow cytometry). There was no statistically significant correlation between day 14 and day 35 BM status (both by morphology and flow cytometry). Sensitivity and specificity of morphological evaluation of BM was much lower compared to minimal residual disease assessment by flow cytometry. There is a need to incorporate flow cytometry in risk stratification of patients who are being treated with BFM 2002 protocol.
Collapse
Affiliation(s)
- Girish Kamat
- 1Department of Hematology, SDM College of Medical Sciences and Hospital, Dharwad, Karnataka India
| | - Kannan Subramanian
- Department of Hematology and Bone Marrow Transplantation, Sahyadri Specialty Hospital, Karve Road, Pune, India
| | - Shashikant Apte
- Department of Hematology and Bone Marrow Transplantation, Sahyadri Specialty Hospital, Karve Road, Pune, India
| |
Collapse
|
11
|
Ampatzidou M, Paterakis G, Vasdekis V, Papadhimitriou SI, Papadakis V, Vassilopoulos G, Polychronopoulou S. Prognostic significance of flow cytometry MRD log reduction during induction treatment of childhood ALL. Leuk Lymphoma 2018; 60:258-261. [PMID: 29963934 DOI: 10.1080/10428194.2018.1471603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Maria Ampatzidou
- a Department of Paediatric Haematology-Oncology , 'Aghia Sophia' Children's Hospital , Athens , Greece
| | - Georgios Paterakis
- b Flow Cytometry Department, Laboratory of Immunology , 'G. Gennimatas' General Hospital , Athens , Greece
| | | | - Stephanos I Papadhimitriou
- d Department of Molecular Cytogenetics, Laboratory of Hematology , 'G. Gennimatas' General Hospital , Athens , Greece
| | - Vassilios Papadakis
- e Department of Hematology , University Hospital of Larisa, Thessaly Medical School , Larisa , Greece
| | - Georgios Vassilopoulos
- e Department of Hematology , University Hospital of Larisa, Thessaly Medical School , Larisa , Greece
| | - Sophia Polychronopoulou
- a Department of Paediatric Haematology-Oncology , 'Aghia Sophia' Children's Hospital , Athens , Greece
| |
Collapse
|
12
|
Next-generation sequencing indicates false-positive MRD results and better predicts prognosis after SCT in patients with childhood ALL. Bone Marrow Transplant 2017; 52:962-968. [PMID: 28244980 DOI: 10.1038/bmt.2017.16] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/19/2016] [Accepted: 01/03/2017] [Indexed: 11/09/2022]
Abstract
Minimal residual disease (MRD) monitoring via quantitative PCR (qPCR) detection of Ag receptor gene rearrangements has been the most sensitive method for predicting prognosis and making post-transplant treatment decisions for patients with ALL. Despite the broad clinical usefulness and standardization of this method, we and others have repeatedly reported the possibility of false-positive MRD results caused by massive B-lymphocyte regeneration after stem cell transplantation (SCT). Next-generation sequencing (NGS) enables precise and sensitive detection of multiple Ag receptor rearrangements, thus providing a more specific readout compared to qPCR. We investigated two cohorts of children with ALL who underwent SCT (30 patients and 228 samples). The first cohort consisted of 17 patients who remained in long-term CR after SCT despite having low MRD positivity (<0.01%) at least once during post-SCT monitoring using qPCR. Only one of 27 qPCR-positive samples was confirmed to be positive by NGS. Conversely, 10 of 15 samples with low qPCR-detected MRD positivity from 13 patients who subsequently relapsed were also confirmed to be positive by NGS (P=0.002). These data show that NGS has a better specificity in post-SCT ALL management and indicate that treatment interventions aimed at reverting impending relapse should not be based on qPCR only.
Collapse
|
13
|
Klener P, Fronkova E, Berkova A, Jaksa R, Lhotska H, Forsterova K, Soukup J, Kulvait V, Vargova J, Fiser K, Prukova D, Alam M, Calvin Lenyeletse Maswabi B, Michalova K, Zemanova Z, Jancuskova T, Pekova S, Trneny M. Mantle cell lymphoma-variant Richter syndrome: Detailed molecular-cytogenetic and backtracking analysis reveals slow evolution of a pre-MCL clone in parallel with CLL over several years. Int J Cancer 2016; 139:2252-60. [DOI: 10.1002/ijc.30263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/18/2016] [Accepted: 06/30/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Pavel Klener
- Department of Hematology; Charles University General Hospital Prague; Prague Czech Republic
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | - Eva Fronkova
- Childhood Leukemia Investigation Prague (CLIP), Faculty Hospital Motol Prague; Prague Czech Republic
| | - Adela Berkova
- Center of Oncocytogenetics, Institute of Clinical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | - Radek Jaksa
- Department of Pathology; Charles University General Hospital Prague; Prague Czech Republic
| | - Halka Lhotska
- Center of Oncocytogenetics, Institute of Clinical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | - Kristina Forsterova
- Department of Hematology; Charles University General Hospital Prague; Prague Czech Republic
| | - Jan Soukup
- Department of Pathology and Molecular Medicine; Second Faculty of Medicine, Faculty Hospital Motol Prague; Prague Czech Republic
| | - Vojtech Kulvait
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | - Jarmila Vargova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | - Karel Fiser
- Childhood Leukemia Investigation Prague (CLIP), Faculty Hospital Motol Prague; Prague Czech Republic
| | - Dana Prukova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | - Mahmudul Alam
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | | | - Kyra Michalova
- Center of Oncocytogenetics, Institute of Clinical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | - Zuzana Zemanova
- Center of Oncocytogenetics, Institute of Clinical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague; Prague Czech Republic
| | | | - Sona Pekova
- Synlab Genetics, Department of Cytogenetics; Prague Czech Republic
| | - Marek Trneny
- Department of Hematology; Charles University General Hospital Prague; Prague Czech Republic
| |
Collapse
|
14
|
Karas-Kuželički N, Mencej-Bedrač S, Jazbec J, Marc J, Mlinarič-Raščan I. Risk factors for symptomatic osteonecrosis in childhood ALL: A retrospective study of a Slovenian pediatric ALL population between 1970 and 2004. Exp Ther Med 2016; 12:840-846. [PMID: 27446285 DOI: 10.3892/etm.2016.3391] [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: 07/27/2015] [Accepted: 04/04/2016] [Indexed: 01/01/2023] Open
Abstract
Treatment induced non-traumatic osteonecrosis (ON) has been reported increasingly in children treated for acute lymphoblastic leukemia (ALL). Several risk factors for ON have been identified in childhood cancer patients; however, their diagnostic and prognostic power is limited and the etiology of the disease remains unclear. Therefore, a continuous effort is focused on the identification of additional ON risk factors. We performed a retrospective study of 313 childhood ALL patients to test the association between the ON occurrence in children receiving ALL therapy and common polymorphisms in potential target genes: Thiopurine S-methyltransferase (TPMT; 460G>A, 719A>G), 5,10-methylenetetrahydrofolate reductase (MTHFR; 677C>T, 1298A>C), estrogen receptor alpha 1 (ESR1; XbaI) and collagen type I, α1 (COL1A1; Sp1). In the present cohort, higher age and more recently developed treatment protocols were independent risk factors for ON. In children >14.5 years old, TPMT genotype modulated the risk of ON. Additionally, in children <12.9 years old ESR1 genotypes were also implicated in the pathogenesis of ON. Besides greater age and more recent treatment protocols, genetic factors (polymorphisms in ESR1 and TPMT genes) were suggested to be implicated in the pathogenesis of ON and could be potentially used as genetic prognostic markers for ON.
Collapse
Affiliation(s)
- Nataša Karas-Kuželički
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Simona Mencej-Bedrač
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Janez Jazbec
- Unit of Oncology and Hematology, University Medical Centre, University Children's Hospital, 1000 Ljubljana, Slovenia
| | - Janja Marc
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Irena Mlinarič-Raščan
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
15
|
Avigad S, Verly IRN, Lebel A, Kordi O, Shichrur K, Ohali A, Hameiri-Grossman M, Kaspers GJL, Cloos J, Fronkova E, Trka J, Luria D, Kodman Y, Mirsky H, Gaash D, Jeison M, Avrahami G, Elitzur S, Gilad G, Stark B, Yaniv I. miR expression profiling at diagnosis predicts relapse in pediatric precursor B-cell acute lymphoblastic leukemia. Genes Chromosomes Cancer 2015; 55:328-39. [PMID: 26684414 DOI: 10.1002/gcc.22334] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 11/11/2022] Open
Abstract
Our aim was to identify miRNAs that can predict risk of relapse in pediatric patients with acute lymphoblastic leukemia (ALL). Following high-throughput miRNA expression analysis (48 samples), five miRs were selected for further confirmation performed by real time quantitative PCR on a cohort of precursor B-cell ALL patients (n = 138). The results were correlated with clinical parameters and outcome. Low expression of miR-151-5p, and miR-451, and high expression of miR-1290 or a combination of all three predicted inferior relapse free survival (P = 0.007, 0.042, 0.025, and <0.0001, respectively). Cox regression analysis identified aberrant expression of the three miRs as an independent prognostic marker with a 10.5-fold increased risk of relapse (P = 0.041) in PCR-MRD non-high risk patients. Furthermore, following exclusion of patients harboring IKZF1 deletion, the aberrant expression of all three miRs could identify patients with a 24.5-fold increased risk to relapse (P < 0.0001). The prognostic relevance of the three miRNAs was evaluated in a non-BFM treated precursor B-cell ALL cohort (n = 33). A significant correlation between an aberrant expression of at least one of the three miRs and poor outcome was maintained (P < 0.0001). Our results identify an expression profile of miR-151-5p, miR-451, and miR-1290 as a novel biomarker for outcome in pediatric precursor B-cell ALL patients, regardless of treatment protocol. The use of these markers may lead to improved risk stratification at diagnosis and allow early therapeutic interventions in an attempt to improve survival of high risk patients.
Collapse
Affiliation(s)
- Smadar Avigad
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iedan R N Verly
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Asaf Lebel
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oshrit Kordi
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Keren Shichrur
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Ohali
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Hameiri-Grossman
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gertjan J L Kaspers
- Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Jacqueline Cloos
- Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Eva Fronkova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University Prague, Czech Republic
| | - Jan Trka
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University Prague, Czech Republic
| | - Drorit Luria
- Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yona Kodman
- Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hadar Mirsky
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dafna Gaash
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marta Jeison
- Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galia Avrahami
- Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sarah Elitzur
- Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gil Gilad
- Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Batia Stark
- Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Isaac Yaniv
- Molecular Oncology, Felsenstein Medical Research Center, Petah Tikva, Israel.,Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
16
|
The predictive strength of next-generation sequencing MRD detection for relapse compared with current methods in childhood ALL. Blood 2015; 126:1045-7. [PMID: 26294720 DOI: 10.1182/blood-2015-07-655159] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
17
|
Kunz JB, Rausch T, Bandapalli OR, Eilers J, Pechanska P, Schuessele S, Assenov Y, Stütz AM, Kirschner-Schwabe R, Hof J, Eckert C, von Stackelberg A, Schrappe M, Stanulla M, Koehler R, Avigad S, Elitzur S, Handgretinger R, Benes V, Weischenfeldt J, Korbel JO, Muckenthaler MU, Kulozik AE. Pediatric T-cell lymphoblastic leukemia evolves into relapse by clonal selection, acquisition of mutations and promoter hypomethylation. Haematologica 2015; 100:1442-50. [PMID: 26294725 DOI: 10.3324/haematol.2015.129692] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/11/2015] [Indexed: 11/09/2022] Open
Abstract
Relapsed precursor T-cell acute lymphoblastic leukemia is characterized by resistance against chemotherapy and is frequently fatal. We aimed at understanding the molecular mechanisms resulting in relapse of T-cell acute lymphoblastic leukemia and analyzed 13 patients at first diagnosis, remission and relapse by whole exome sequencing, targeted ultra-deep sequencing, multiplex ligation dependent probe amplification and DNA methylation array. Compared to primary T-cell acute lymphoblastic leukemia, in relapse the number of single nucleotide variants and small insertions and deletions approximately doubled from 11.5 to 26. Targeted ultra-deep sequencing sensitively detected subclones that were selected for in relapse. The mutational pattern defined two types of relapses. While both are characterized by selection of subclones and acquisition of novel mutations, 'type 1' relapse derives from the primary leukemia whereas 'type 2' relapse originates from a common pre-leukemic ancestor. Relapse-specific changes included activation of the nucleotidase NT5C2 resulting in resistance to chemotherapy and mutations of epigenetic modulators, exemplified by SUZ12, WHSC1 and SMARCA4. While mutations present in primary leukemia and in relapse were enriched for known drivers of leukemia, relapse-specific changes revealed an association with general cancer-promoting mechanisms. This study thus identifies mechanisms that drive progression of pediatric T-cell acute lymphoblastic leukemia to relapse and may explain the characteristic treatment resistance of this condition.
Collapse
Affiliation(s)
- Joachim B Kunz
- Department of Pediatric Oncology, Hematology and Immunology, Children's Hospital, University of Heidelberg, Germany Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Tobias Rausch
- Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany European Molecular Biology Laboratory (EMBL), Genomics Core Facility, Heidelberg, Germany
| | - Obul R Bandapalli
- Department of Pediatric Oncology, Hematology and Immunology, Children's Hospital, University of Heidelberg, Germany Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Juliane Eilers
- Department of Pediatric Oncology, Hematology and Immunology, Children's Hospital, University of Heidelberg, Germany Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany
| | - Paulina Pechanska
- Department of Pediatric Oncology, Hematology and Immunology, Children's Hospital, University of Heidelberg, Germany Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany
| | - Stephanie Schuessele
- Department of Pediatric Oncology, Hematology and Immunology, Children's Hospital, University of Heidelberg, Germany Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany
| | - Yassen Assenov
- Division of Epigenomics and Cancer Risk Factors, The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adrian M Stütz
- Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | | | - Jana Hof
- Department of Pediatric Oncology/Hematology, Charité - Universitätsmedizin Berlin, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cornelia Eckert
- Department of Pediatric Oncology/Hematology, Charité - Universitätsmedizin Berlin, Germany
| | - Arend von Stackelberg
- Department of Pediatric Oncology/Hematology, Charité - Universitätsmedizin Berlin, Germany
| | - Martin Schrappe
- Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Martin Stanulla
- Department of Pediatric Hematology/Oncology, Medical School Hannover, Germany
| | - Rolf Koehler
- Department of Human Genetics, University of Heidelberg, Germany
| | - Smadar Avigad
- Molecular Oncology, Felsenstein Medical Research Center and Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Sarah Elitzur
- Molecular Oncology, Felsenstein Medical Research Center and Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | | | - Vladimir Benes
- European Molecular Biology Laboratory (EMBL), Genomics Core Facility, Heidelberg, Germany
| | - Joachim Weischenfeldt
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Jan O Korbel
- Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, Children's Hospital, University of Heidelberg, Germany Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany
| | - Andreas E Kulozik
- Department of Pediatric Oncology, Hematology and Immunology, Children's Hospital, University of Heidelberg, Germany Molecular Medicine Partnership Unit, EMBL-University of Heidelberg, Germany German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
18
|
Wei W, Chen X, Zou Y, Chang L, An W, Wan Y, Liu T, Yang W, Chen Y, Guo Y, Zhu X. Prediction of outcomes by early treatment responses in childhood T-cell acute lymphoblastic leukemia: a retrospective study in China. BMC Pediatr 2015; 15:80. [PMID: 26174476 PMCID: PMC4502910 DOI: 10.1186/s12887-015-0390-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/17/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Early treatment responses are important prognostic factors in childhood T-cell acute lymphoblastic leukemia (T-ALL) patients. The predictive values of early treatment responses in Chinese childhood T-ALL patients were still unknown. METHODS From January 2003 to December 2012, 74 consecutive patients aged ≤ 15 years with newly diagnosed T-ALL were treated with BCH-2003 protocol or CCLG-2008 protocol in the Department of Pediatric, Institute of Hematology and Blood Diseases Hospital in China. Predictive values of early treatment responses, including prednisone response, bone marrow morphology at day 15 and day 33 during induction chemotherapy, and minimal residual disease (MRD) monitored by flow cytometry after induction therapy (time point 1, TP1) and before consolidation therapy (time point 2, TP2), were analyzed. RESULTS The 5-year event free survival (EFS) and overall survival (OS) rates for these patients were 62.5% (SE, 6.4) and 62.7% (SE, 6.6), respectively. Prednisone poor responder was strongly associated with increased chance of induction failure (14.8%) and decreased survival rate (5 year EFS rate, 51.1 % (SE, 10.5)). Patients with ≥ 25% blast cells in bone marrow at day 15 were more likely to have an inferior outcome. 93.2% of the T-ALL patients achieved complete remission at day 33 while patients with resistant disease all died of disease progression. MRD ≥ 10(-2) at TP1 or MRD ≥ 10(-3) at TP2 was significantly related to dismal prognosis. Risk groups classified by MRD at two time points could stratify patients into different groups: 29.0% of the patients were MRD standard risk (MRD < 10(-4) at both time points) with 3-year EFS rate of 100%, 29.0% were MRD high risk (MRD ≥ 10(-2) at TP1 or MRD ≥ 10(-2) at TP2) with 3-year EFS rate of 55.6% (SE, 16.6) , and the rest of patients were defined as MRD intermediate risk with 3-year EFS rate of 85.7% (SE, 13.2). CONCLUSION Our study demonstrated that MRD was the most powerful predictor of treatment outcome in childhood T-ALL patients and conventional morphological assessments of treatment response still played important roles in predicting treatment outcome and tailoring treatment intensity especially in countries with inadequate skills or financial resources for MRD monitoring.
Collapse
Affiliation(s)
- Wei Wei
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Xiaojuan Chen
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Yao Zou
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Lixian Chang
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Wenbin An
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Yang Wan
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Tianfeng Liu
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Wenyu Yang
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Yumei Chen
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Ye Guo
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| | - Xiaofan Zhu
- Department of Pediatric, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, Peoples Republic of China.
| |
Collapse
|
19
|
Schrappe M. Detection and management of minimal residual disease in acute lymphoblastic leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:244-249. [PMID: 25696862 DOI: 10.1182/asheducation-2014.1.244] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The detection of minimal residual disease (MRD) has become part of the state-of-the-art diagnostics to guide treatment both in pediatric and adult acute lymphoblastic leukemia (ALL). This applies to the treatment of de novo and recurrent ALL. In high-risk ALL, MRD detection is considered an important tool to adjust therapy before and after hematopoietic stem cell transplantation. Precise quantification and quality control is instrumental to avoid false treatment assignment. A new methodological approach to analyzing MRD has become available and is based on next-generation sequencing. In principle, this technique will be able to detect a large number of leukemic subclones at a much higher speed than before. Carefully designed prospective studies need to demonstrate concordance or even superiority compared with those techniques in use right now: detection of aberrant expression of leukemia-specific antigens by flow cytometry of blood or bone marrow, or detection of specific rearrangements of the T-cell receptor or immunoglobulin genes by real-time quantitative polymerase chain reaction using DNA of leukemic cells. In some cases with known fusion genes, such as BCR/ABL, reverse transcriptase-polymerase chain reaction has been used as additional method to identify leukemic cells by analyzing RNA in patient samples. MRD detection may be used to modulate treatment intensity once it has been demonstrated at well-defined informative checkpoints that certain levels of MRD can reliably predict the risk of relapse. In addition, MRD is used as end point to determine the activity of a given agent or treatment protocol. If activity translates into antileukemic efficacy, MRD may be considered a surrogate clinical end point.
Collapse
Affiliation(s)
- Martin Schrappe
- Department of Pediatrics, Christian-Albrechts-University of Kiel, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
20
|
Host genetic variants of ABCB1 and IL15 influence treatment outcome in paediatric acute lymphoblastic leukaemia. Br J Cancer 2014; 110:1673-80. [PMID: 24434428 PMCID: PMC3960629 DOI: 10.1038/bjc.2014.7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 12/11/2013] [Accepted: 12/17/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Host germline variations and their potential prognostic importance is an emerging area of interest in paediatric ALL. METHODS We investigated the associations between 20 germline variations and various clinical end points in 463 children with ALL. RESULTS After adjusting for known prognostic factors, variants in two genes were found to be independently associated with poorer EFS: ABCB1 T/T at either 2677 (rs2032582) or 3435 (rs1045642) position (P=0.003) and IL15 67276493G/G (rs17015014; P=0.022). These variants showed a strong additive effect affecting outcome (P<0.001), whereby patients with both risk genotypes had the worst EFS (P=0.001), even after adjusting for MRD levels at the end of remission induction. The adverse effect of ABCB1 T/T genotypes was most pronounced in patients with favourable cytogenetics (P=0.011) while the IL15 67276493G/G genotype mainly affected patients without common chromosomal abnormalities (P=0.022). In both cytogenetic subgroups, increasing number of such risk genotypes still predicted worsening outcome (P<0.001 and=0.009, respectively). CONCLUSION These results point to the prognostic importance of host genetic variants, although the specific mechanisms remain unclarified. Inclusion of ABCB1 and IL15 variants may help improve risk assignment strategies in paediatric ALL.
Collapse
|
21
|
Abstract
SUMMARY Predictive/prognostic factors in acute leukemia continue to be sought, in order to refine treatment strategies. Minimal residual disease (MRD) testing has been shown to be a statistically significant factor by multivariate analysis in both acute lymphoblastic leukemia (ALL) and acute myeloid leukemia. Its utility in guiding therapy has been more extensively studied in pediatric ALL, with some protocols having instituted MRD testing into therapeutic algorithms. The clinical impact of MRD testing in ALL and acute myeloid leukemia will be presented, including both molecular and flow cytometric methodologies, with a more focused discussion of the strategy, methodology and interpretation of MRD testing by multiparametric flow cytometry.
Collapse
Affiliation(s)
- Lorinda Soma
- University of Washington, Department of Laboratory Medicine, Division of Hematopathology, Room NW120, Box 357110, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Brent Wood
- University of Washington, Department of Laboratory Medicine, Division of Hematopathology, Room NW120, Box 357110, 1959 NE Pacific Street, Seattle, WA 98195, USA
| |
Collapse
|
22
|
CD2-positive B-cell precursor acute lymphoblastic leukemia with an early switch to the monocytic lineage. Leukemia 2013; 28:609-20. [PMID: 24270736 DOI: 10.1038/leu.2013.354] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/10/2013] [Accepted: 11/14/2013] [Indexed: 12/20/2022]
Abstract
Switches from the lymphoid to myeloid lineage during B-cell precursor acute lymphoblastic leukemia (BCP-ALL) treatment are considered rare and thus far have been detected in MLL-rearranged leukemia. Here, we describe a novel BCP-ALL subset, switching BCP-ALL or swALL, which demonstrated monocytosis early during treatment. Despite their monocytic phenotype, 'monocytoids' share immunoreceptor gene rearrangements with leukemic B lymphoblasts. All swALLs demonstrated BCP-ALL with CD2 positivity and no MLL alterations, and the proportion of swALLs cases among BCP-ALLs was unexpectedly high (4%). The upregulation of CEBPα and demethylation of the CEBPA gene were significant in blasts at diagnosis, prior to the time when most of the switching occurs. Intermediate stages between CD14(neg)CD19(pos)CD34(pos) B lymphoblasts and CD14(pos)CD19(neg)CD34(neg) 'monocytoids' were detected, and changes in the expression of PAX5, PU1, M-CSFR, GM-CSFR and other genes accompanied the switch. Alterations in the Ikaros and ERG genes were more frequent in swALL patients; however, both were altered in only a minority of swALLs. Moreover, switching could be recapitulated in vitro and in mouse xenografts. Although children with swALL respond slowly to initial therapy, risk-based ALL therapy appears the treatment of choice for swALL. SwALL shows that transdifferentiating into monocytic lineage is specifically associated with CEBPα changes and CD2 expression.
Collapse
|
23
|
Yeoh AEJ, Tan D, Li CK, Hori H, Tse E, Pui CH. Management of adult and paediatric acute lymphoblastic leukaemia in Asia: resource-stratified guidelines from the Asian Oncology Summit 2013. Lancet Oncol 2013; 14:e508-23. [PMID: 24176570 PMCID: PMC4059516 DOI: 10.1016/s1470-2045(13)70452-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Survival for adults and children with acute lymphoblastic leukaemia has risen substantially in recent years because use of improved risk-directed treatments and supportive care has widened. In nearly all developed countries, multidisciplinary panels of leukaemia experts have formulated clinical practice guidelines in which standard treatment approaches are recommended on the basis of current evidence. However, those guidelines do not take into account resource limitations in low-income countries, including financial and technical challenges. In Asia, huge disparities in economy and infrastructure exist between countries, and even among different regions in some large countries. At a consensus session held as part of the 2013 Asian Oncology Summit in Bangkok, Thailand, a panel of experts summarised recommendations for management of adult and paediatric acute lymphoblastic leukaemia. Strategies were developed for Asian countries on the basis of available financial, skill, and logistical resources and were stratified in a four-tier system according to the resources available in a particular country or region (basic, limited, enhanced, and maximum).
Collapse
Affiliation(s)
- Allen E J Yeoh
- Department of Paediatrics, National University Hospital, National University of Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
24
|
Volejnikova J, Mejstrikova E, Dörge P, Meissner B, Zimmermannova O, Svojgr K, Stanulla M, Cario G, Schrappe M, Stary J, Hrusak O, Trka J, Fronkova E. Ikaros (IKZF1) alterations and minimal residual disease at day 15 assessed by flow cytometry predict prognosis of childhood BCR/ABL-negative acute lymphoblastic leukemia. Pediatr Blood Cancer 2013; 60:420-7. [PMID: 22997141 DOI: 10.1002/pbc.24299] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 08/06/2012] [Indexed: 11/10/2022]
Abstract
BACKGROUND Recently, several studies have demonstrated a negative prognostic impact of Ikaros (IKZF1) gene alterations in acute lymphoblastic leukemia (ALL). However, controversies still exist regarding the impact of IKZF1 in current treatment protocols. PROCEDURE We simultaneously detected IKZF1 gene deletions by multiplex ligation-dependent probe amplification and gene expression of IKZF1 isoforms in 206 children with BCR/ABL-negative ALL treated with ALL IC-BFM 2002 protocol, in which risk stratification was not based on minimal residual disease (MRD), and validated the results on a cohort of 189 patients treated with MRD-directed ALL-BFM 2000 protocol. RESULTS Deletion of IKZF1 was present in 14 of 206 (7%) ALL IC patients. Interestingly, gene expression did not completely correlate with the deletion status in either cohort. Deletions were not always reflected in the gene expression of dominant-negative isoforms, and conversely, 7 of 395 (2%) non-deleted cases overexpressed dominant-negative isoform Ik6. IKZF1 deletions significantly affected event-free survival (EFS) of the ALL IC cohort (41 ± 14% vs. 86 ± 3%, P < 0.0001). Regarding IKZF1 isoforms, only Ik6 overexpression had negative prognostic impact (EFS 50 ± 16% vs. 85 ± 3%, P = 0.003). In multivariate analysis, which included ALL IC risk criteria, flow-cytometric MRD and IKZF1 alterations, day 15 MRD and IKZF1 deletion status displayed an independent prognostic impact. CONCLUSIONS We show that MRD-directed treatment diminishes prognostic impact of IKZF1 alterations. However, IKZF1 status alone or combined with day 15 flow cytometry can significantly improve risk stratification within BFM protocols at centers that do not perform antigen-receptor-based MRD monitoring.
Collapse
Affiliation(s)
- Jana Volejnikova
- 2nd Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University and University Hospital Motol, Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cheng SH, Lau KM, Li CK, Chan NPH, Ip RKL, Cheng CK, Lee V, Shing MMK, Leung AWK, Ha SY, Cheuk DKL, Lee ACW, Li CH, Luk CW, Ling SC, Hrusak O, Mejstrikova E, Leung Y, Ng MHL. Minimal residual disease-based risk stratification in Chinese childhood acute lymphoblastic leukemia by flow cytometry and plasma DNA quantitative polymerase chain reaction. PLoS One 2013; 8:e69467. [PMID: 23936021 PMCID: PMC3723913 DOI: 10.1371/journal.pone.0069467] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 06/10/2013] [Indexed: 02/05/2023] Open
Abstract
Minimal residual disease, or MRD, is an important prognostic indicator in childhood acute lymphoblastic leukemia. In ALL-IC-BFM 2002 study, we employed a standardized method of flow cytometry MRD monitoring for multiple centers internationally using uniformed gating, and determined the relevant MRD-based risk stratification strategies in our local patient cohort. We also evaluated a novel method of PCR MRD quantitation using peripheral blood plasma. For the bone marrow flow MRD study, patients could be stratified into 3 risk groups according to MRD level using a single time-point at day-15 (Model I) (I-A: <0.1%, I-B: 0.1-10%, I-C: >10%), or using two time-points at day-15 and day-33 (Model II) (II-A: day-15<10% and day-33<0.01%, II-B: day-15 ≥ 10% or day-33 ≥ 0.01% but not both, II-C: day-15 ≥ 10% and day-33 ≥ 0.01%), which showed significantly superior prediction of relapse (p = .00047 and <0.0001 respectively). Importantly, patients with good outcome (frequency: 56.0%, event-free survival: 90.1%) could be more accurately predicted by Model II. In peripheral blood plasma PCR MRD investigation, patients with day-15-MRD ≥ 10(-4) were at a significantly higher risk of relapse (p = 0.0117). By multivariate analysis, MRD results from both methods could independently predict patients' prognosis, with 20-35-fold increase in risk of relapse for flow MRD I-C and II-C respectively, and 5.8-fold for patients having plasma MRD of ≥ 10(-4). We confirmed that MRD detection by flow cytometry is useful for prognostic evaluation in our Chinese cohort of childhood ALL after treatment. Moreover, peripheral blood plasma DNA MRD can be an alternative where bone marrow specimen is unavailable and as a less invasive method, which allows close monitoring.
Collapse
Affiliation(s)
- Suk Hang Cheng
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Kin Mang Lau
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Kong Li
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Natalie P. H. Chan
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Rosalina K. L. Ip
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Keung Cheng
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Vincent Lee
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Matthew M. K. Shing
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Alex W. K. Leung
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Shau Yin Ha
- Department of Pediatrics, Queen Mary Hospital, Hong Kong, China
| | | | | | - Chak Ho Li
- Department of Pediatrics, Tuen Mun Hospital, Hong Kong, China
| | - Chung Wing Luk
- Department of Pediatrics, Queen Elizabeth Hospital, Hong Kong, China
| | - Siu Cheung Ling
- Department of Pediatrics, Princess Margaret Hospital, Hong Kong, China
| | - Ondrej Hrusak
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ester Mejstrikova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Yonna Leung
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Margaret H. L. Ng
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong, China
- * E-mail:
| |
Collapse
|
26
|
Abstract
Abstract
After approximately 20 years of development and after several prospective clinical trials, the detection of minimal residual disease (MRD) has emerged as part of state-of-the-art diagnostics to guide the majority of contemporary treatment programs both in pediatric and adult acute lymphoblastic leukemia (ALL). For ALL, several methods of MRD analysis are available, but 2 are widely applicable. One is based on the detection of aberrant expression of leukemia specific antigens by flow cytometry and the other one uses the specific rearrangements of the TCR or Ig genes, which can be detected by quantitative PCR in the DNA of leukemic cells. In some cases with known fusion genes such as BCR/ABL, RT-PCR can be used as a third method of identifying leukemic cells by analyzing RNA in patient samples. Clinical application of such sophisticated tools in the stratification and treatment of ALL requires reliable, reproducible, and quality-assured methods to ensure patient safety.
Collapse
|
27
|
Marjerrison S, Antillon F, Fu L, Martinez R, Vasquez R, Bonilla M, Howard SC, Sung L. Outcome of children treated for relapsed acute lymphoblastic leukemia in Central America. Cancer 2012; 119:1277-83. [DOI: 10.1002/cncr.27846] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 08/16/2012] [Accepted: 09/10/2012] [Indexed: 11/07/2022]
|
28
|
Zhai X, Wang H, Zhu X, Miao H, Qian X, Li J, Gao Y, Lu F, Wu Y. Gene polymorphisms of ABC transporters are associated with clinical outcomes in children with acute lymphoblastic leukemia. Arch Med Sci 2012; 8:659-71. [PMID: 23056078 PMCID: PMC3460503 DOI: 10.5114/aoms.2012.30290] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 04/16/2012] [Accepted: 05/11/2012] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Genetic variability affects clinical outcome in pediatric acute lymphocytic leukemia (ALL) patients. Evaluating gene polymorphisms in ABC transporters could help identify relapse risk and predict outcome. MATERIAL AND METHODS The SNaPshot SNP technique was used to analyze single-nucleotide polymorphisms (SNPs) in the multidrug transporter 1 (MDR1), multidrug resistance associated proteins (MRP1, MRP2) and breast cancer resistance protein (BCRP) genes of 82 pediatric ALL patients. The association between the SNPs with risk of all events and death as well as with survival was evaluated by the univariate Cox proportional hazard model. RESULTS The BCRP G34A SNP was the only SNP significantly associated with ALL. Risk factors included pre-treatment WBC counts and post-treatment peripheral and bone marrow leukemic cell counts. We found no association between MDR1 SNPs with these factors. The BCRP C421A C/A and C/C genotypes were significantly associated with low pre-treatment WBC counts while MRP2 G1249A G/G was significantly associated with low levels of post-treatment peripheral and bone marrow leukemic cells. A combination of C1236T, G1249A and/or G34A SNPs was significantly associated with lower EFS and OS. CONCLUSIONS Polymorphisms associated with risk of ALL and clinical outcome may be potential biomarkers to predict clinical outcome and improve prognosis in childhood ALL.
Collapse
Affiliation(s)
- Xiaowen Zhai
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Hongsheng Wang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Xiaohua Zhu
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Hui Miao
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Xiaowen Qian
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Jun Li
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Yijin Gao
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Fengjuan Lu
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Yue Wu
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, P.R. China
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, P.R. China
| |
Collapse
|
29
|
MLPA is a powerful tool for detecting lymphoblastic transformation in chronic myeloid leukemia and revealing the clonal origin of relapse in pediatric acute lymphoblastic leukemia. Cancer Genet 2012; 205:465-9. [DOI: 10.1016/j.cancergen.2012.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 05/10/2012] [Accepted: 05/14/2012] [Indexed: 10/27/2022]
|
30
|
Gavidia R, Fuentes SL, Vasquez R, Bonilla M, Ethier MC, Diorio C, Caniza M, Howard SC, Sung L. Low socioeconomic status is associated with prolonged times to assessment and treatment, sepsis and infectious death in pediatric fever in El Salvador. PLoS One 2012; 7:e43639. [PMID: 22928008 PMCID: PMC3425537 DOI: 10.1371/journal.pone.0043639] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 07/23/2012] [Indexed: 11/18/2022] Open
Abstract
Background Infection remains the most common cause of death from toxicity in children with cancer in low- and middle-income countries. Rapid administration of antibiotics when fever develops can prevent progression to sepsis and shock, and serves as an important indicator of the quality of care in children with acute lymphoblastic leukemia and acute myeloid leukemia. We analyzed factors associated with (1) Longer times from fever onset to hospital presentation/antibiotic treatment and (2) Sepsis and infection-related mortality. Method This prospective cohort study included children aged 0–16 years with newly diagnosed acute leukemia treated at Benjamin Bloom Hospital, San Salvador. We interviewed parents/caregivers within one month of diagnosis and at the onset of each new febrile episode. Times from initial fever to first antibiotic administration and occurrence of sepsis and infection-related mortality were documented. Findings Of 251 children enrolled, 215 had acute lymphoblastic leukemia (85.7%). Among 269 outpatient febrile episodes, median times from fever to deciding to seek medical care was 10.0 hours (interquartile range [IQR] 5.0–20.0), and from decision to seek care to first hospital visit was 1.8 hours (IQR 1.0–3.0). Forty-seven (17.5%) patients developed sepsis and 7 (2.6%) died of infection. Maternal illiteracy was associated with longer time from fever to decision to seek care (P = 0.029) and sepsis (odds ratio [OR] 3.06, 95% confidence interval [CI] 1.09–8.63; P = 0.034). More infectious deaths occurred in those with longer travel time to hospital (OR 1.36, 95% CI 1.03–1.81; P = 0.031) and in families with an annual household income <US$2,000 (OR 13.90, 95% CI 1.62–119.10; P = 0.016). Interpretation Illiteracy, poverty, and longer travel times are associated with delays in assessment and treatment of fever and with sepsis and infectious mortality in pediatric leukemia. Providing additional education to high-risk families and staying at a nearby guest house during periods of neutropenia may decrease sepsis and infectious mortality.
Collapse
Affiliation(s)
- Ronald Gavidia
- Pediatric Oncology, Benjamin Bloom National Children’s Hospital, San Salvador, El Salvador
| | - Soad L. Fuentes
- Pediatric Oncology, Benjamin Bloom National Children’s Hospital, San Salvador, El Salvador
| | - Roberto Vasquez
- Pediatric Oncology, Benjamin Bloom National Children’s Hospital, San Salvador, El Salvador
| | - Miguel Bonilla
- Pediatric Oncology, Benjamin Bloom National Children’s Hospital, San Salvador, El Salvador
| | - Marie-Chantal Ethier
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Canada
| | - Caroline Diorio
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Canada
| | - Miguela Caniza
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- International Outreach Program, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Scott C. Howard
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- International Outreach Program, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Lillian Sung
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Canada
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- * E-mail:
| |
Collapse
|
31
|
CD105 and placental growth factor – Potent prognostic factors in childhood acute lymphoblastic leukaemia. Leuk Res 2012; 36:846-51. [DOI: 10.1016/j.leukres.2012.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 02/27/2012] [Accepted: 03/18/2012] [Indexed: 11/20/2022]
|
32
|
Kotur N, Stankovic B, Kassela K, Georgitsi M, Vicha A, Leontari I, Dokmanovic L, Janic D, Krstovski N, Klaassen K, Radmilovic M, Stojiljkovic M, Nikcevic G, Simeonidis A, Sivolapenko G, Pavlovic S, Patrinos GP, Zukic B. 6-mercaptopurine influences TPMT gene transcription in a TPMT gene promoter variable number of tandem repeats-dependent manner. Pharmacogenomics 2012; 13:283-95. [DOI: 10.2217/pgs.11.153] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Aim: TPMT activity is characterized by a trimodal distribution, namely low, intermediate and high methylator. TPMT gene promoter contains a variable number of GC-rich tandem repeats (VNTRs), namely A, B and C, ranging from three to nine repeats in length in an AnBmC architecture. We have previously shown that the VNTR architecture in the TPMT gene promoter affects TPMT gene transcription. Materials, methods & results: Here we demonstrate, using reporter assays, that 6-mercaptopurine (6-MP) treatment results in a VNTR architecture-dependent decrease of TPMT gene transcription, mediated by the binding of newly recruited protein complexes to the TPMT gene promoter, upon 6-MP treatment. We also show that acute lymphoblastic leukemia patients undergoing 6-MP treatment display a VNTR architecture-dependent response to 6-MP. Conclusion: These data suggest that the TPMT gene promoter VNTR architecture can be potentially used as a pharmacogenomic marker to predict toxicity due to 6-MP treatment in acute lymphoblastic leukemia patients. Original submitted 27 July 2011; Revision submitted 24 October 2011
Collapse
Affiliation(s)
- Nikola Kotur
- Institute of Molecular Genetics & Genetic Engineering, University of Belgrade, Laboratory for Molecular Hematology, Belgrade, 11010, Serbia
| | - Biljana Stankovic
- Institute of Molecular Genetics & Genetic Engineering, University of Belgrade, Laboratory for Molecular Hematology, Belgrade, 11010, Serbia
| | - Katerina Kassela
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, GR-26504, Greece
| | - Marianthi Georgitsi
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, GR-26504, Greece
| | - Anna Vicha
- Hematology Division, School of Health Sciences, Faculty of Medicine, University of Patras, Patras, GR-26504, Greece
| | - Iliana Leontari
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, GR-26504, Greece
| | - Lidija Dokmanovic
- University Children’s Hospital, School of Medicine, University of Belgrade, Belgrade, 11000, Serbia
| | - Dragana Janic
- University Children’s Hospital, School of Medicine, University of Belgrade, Belgrade, 11000, Serbia
| | - Nada Krstovski
- University Children’s Hospital, School of Medicine, University of Belgrade, Belgrade, 11000, Serbia
| | - Kristel Klaassen
- Institute of Molecular Genetics & Genetic Engineering, University of Belgrade, Laboratory for Molecular Hematology, Belgrade, 11010, Serbia
| | - Milena Radmilovic
- Institute of Molecular Genetics & Genetic Engineering, University of Belgrade, Laboratory for Molecular Hematology, Belgrade, 11010, Serbia
| | - Maja Stojiljkovic
- Institute of Molecular Genetics & Genetic Engineering, University of Belgrade, Laboratory for Molecular Hematology, Belgrade, 11010, Serbia
| | - Gordana Nikcevic
- Institute of Molecular Genetics & Genetic Engineering, University of Belgrade, Laboratory for Molecular Hematology, Belgrade, 11010, Serbia
| | - Argiris Simeonidis
- Hematology Division, School of Health Sciences, Faculty of Medicine, University of Patras, Patras, GR-26504, Greece
| | - Gregory Sivolapenko
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, GR-26504, Greece
| | - Sonja Pavlovic
- Institute of Molecular Genetics & Genetic Engineering, University of Belgrade, Laboratory for Molecular Hematology, Belgrade, 11010, Serbia
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, GR-26504, Greece
| | - Branka Zukic
- Institute of Molecular Genetics & Genetic Engineering, University of Belgrade, Laboratory for Molecular Hematology, Belgrade, 11010, Serbia
| |
Collapse
|
33
|
Brüggemann M, Gökbuget N, Kneba M. Acute Lymphoblastic Leukemia: Monitoring Minimal Residual Disease as a Therapeutic Principle. Semin Oncol 2012; 39:47-57. [DOI: 10.1053/j.seminoncol.2011.11.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
34
|
Lauten M, Möricke A, Beier R, Zimmermann M, Stanulla M, Meissner B, Odenwald E, Attarbaschi A, Niemeyer C, Niggli F, Riehm H, Schrappe M. Prediction of outcome by early bone marrow response in childhood acute lymphoblastic leukemia treated in the ALL-BFM 95 trial: differential effects in precursor B-cell and T-cell leukemia. Haematologica 2012; 97:1048-56. [PMID: 22271901 DOI: 10.3324/haematol.2011.047613] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND In the ALL-BFM 95 trial for treatment of acute lymphoblastic leukemia, response to a prednisone pre-phase (prednisone response) was used for risk stratification in combination with age and white blood cell count at diagnosis, response to induction therapy and specific genetic high-risk features. DESIGN AND METHODS Cytomorphological marrow response was prospectively assessed on Day 15 during induction, and its prognostic value was analyzed in 1,431 patients treated on ALL-BFM 95. RESULTS The 8-year probabilities of event-free survival were 86.1%, 74.5%, and 46.4% for patients with M1, M2, and M3 Day 15 marrows, respectively. Compared to prednisone response, Day 15 marrow response was superior in outcome prediction in precursor B-cell and T-cell leukemia with, however, a differential effect depending on blast lineage. Outcome was poor in T-cell leukemia patients with prednisone poor-response independent of Day 15 marrow response, whereas among patients with prednisone good-response different risk groups could be identified by Day 15 marrow response. In contrast, prednisone response lost prognostic significance in precursor B-cell leukemia when stratified by Day 15 marrow response. Age and white blood cell count retained their independent prognostic effect. CONCLUSIONS Selective addition of Day 15 marrow response to conventional stratification criteria applied on ALL-BFM 95 (currently in use in several countries as regular chemotherapy protocol for childhood acute lymphoblastic leukemia) may significantly improve risk-adapted treatment delivery. Even though cutting-edge trial risk stratification is meanwhile dominated by minimal residual disease evaluation, an improved conventional risk assessment, as presented here, could be of great importance to countries that lack the technical and/or financial resources associated with the application of minimal residual disease analysis.
Collapse
Affiliation(s)
- Melchior Lauten
- Pediatric Hematology and Oncology, University Hospital Schleswig-Holstein, Lübeck Campus, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Alonso CN, Gallego MS, Rossi JG, Medina A, Rubio PL, Bernasconi AR, Zubizarreta PA, Felice MS. RT-PCR diagnosis of recurrent rearrangements in pediatric acute lymphoblastic leukemia in Argentina. Leuk Res 2012; 36:704-8. [PMID: 22226019 DOI: 10.1016/j.leukres.2011.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/15/2011] [Accepted: 12/07/2011] [Indexed: 10/14/2022]
Abstract
The present study was performed to establish the prevalence of the recurrent fusion transcripts in Argentinean pediatric patients with acute lymphoblastic leukemia (ALL). A total of 380 newly diagnosed children (including 50 infants and 44 T-ALL) were screened by RT-PCR; the incidence of recurrent rearrangements was: ETV6-RUNX1, 12.9%; TCF3-PBX1, 5.0%; BCR-ABL1, 1.6%; and MLL rearrangements, 10.5%. STIL-TAL1 was detected in 22.7% of T-ALL cases. In B-ALL cases, the pEFS was significantly influenced by the presence of genetic alterations. RT-PCR studies improved patients' stratification and also the overall outcome of children treated in a pediatric hospital from a developing country.
Collapse
Affiliation(s)
- Cristina N Alonso
- Department of Hematology and Oncology, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
History of treatment and long-term outcome in children with acute lymphoblastic leukemia in the Czech Republic. MEMO-MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2011. [DOI: 10.1007/s12254-011-0289-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
37
|
Volejnikova J, Mejstrikova E, Valova T, Reznickova L, Hodonska L, Mihal V, Sterba J, Jabali Y, Prochazkova D, Blazek B, Hak J, Cerna Z, Hrusak O, Stary J, Trka J, Fronkova E. Minimal residual disease in peripheral blood at day 15 identifies a subgroup of childhood B-cell precursor acute lymphoblastic leukemia with superior prognosis. Haematologica 2011; 96:1815-21. [PMID: 21880630 DOI: 10.3324/haematol.2011.042937] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Most minimal residual disease-directed treatment interventions in current treatment protocols for acute lymphoblastic leukemia are based on bone marrow testing, which is a consequence of previous studies showing the superiority of bone marrow over peripheral blood as an investigational material. Those studies typically did not explore the prognostic impact of peripheral blood involvement and lacked samples from very early time points of induction. DESIGN AND METHODS In this study, we employed real-time quantitative polymerase chain reaction analysis to examine minimal residual disease in 398 pairs of blood and bone marrow follow-up samples taken from 95 children with B-cell precursor acute lymphoblastic leukemia treated with the ALL IC-BFM 2002 protocol. RESULTS We confirmed the previously published poor correlation between minimal residual disease in blood and marrow at early treatment time points, with levels in bone marrow being higher than in blood in most samples (median 7.9-fold, range 0.04-8,293-fold). A greater involvement of peripheral blood at diagnosis was associated with a higher white blood cell count at diagnosis (P=0.003) and with enlargement of the spleen (P=0.0004) and liver (P=0.05). At day 15, a level of minimal residual disease in blood lower than 10(-4) was associated with an excellent 5-year relapse-free survival in 78 investigated patients (100% versus 69 ± 7%; P=0.0003). Subgroups defined by the level of minimal residual disease in blood at day 15 (high-risk: ≥ 10(-2), intermediate-risk: <10(-2) and ≥ 10(-4), standard-risk: <10(-4)) partially correlated with bone marrow-based stratification described previously, but the risk groups did not match completely. No other time point analyses were predictive of outcome in peripheral blood, except for a weak association at day 8. CONCLUSIONS Minimal residual disease in peripheral blood at day 15 identified a large group of patients with an excellent prognosis and added prognostic information to the risk stratification based on minimal residual disease at day 33 and week 12.
Collapse
Affiliation(s)
- Jana Volejnikova
- Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: results of the AIEOP-BFM-ALL 2000 study. Blood 2011; 118:2077-84. [DOI: 10.1182/blood-2011-03-338707] [Citation(s) in RCA: 296] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Abstract
The prognostic value of MRD in large series of childhood T-ALL has not yet been established. Trial AIEOP-BFM-ALL 2000 introduced standardized quantitative assessment of MRD for stratification, based on immunoglobulin and TCR gene rearrangements as polymerase chain reaction targets: Patients were considered MRD standard risk (MRD-SR) if MRD was negative at day 33 (time point 1 [TP1]) and day 78 (TP2), analyzed by at least 2 sensitive markers; MRD intermediate risk (MRD-IR) if positive either at day 33 or 78 and < 10−3 at day 78; and MRD high risk (MRD-HR) if ≥ 10−3 at day 78. A total of 464 patients with T-ALL were stratified by MRD: 16% of them were MRD-SR, 63% MRD-IR, and 21% MRD-HR. Their 7-year event-free-survival (SE) was 91.1% (3.5%), 80.6% (2.3%), and 49.8% (5.1%) (P < .001), respectively. Negativity of MRD at TP1 was the most favorable prognostic factor. An excellent outcome was also obtained in 32% of patients turning MRD negative only at TP2, indicating that early (TP1) MRD levels were irrelevant if MRD at TP2 was negative (48% of all patients). MRD ≥ 10−3 at TP2 constitutes the most important predictive factor for relapse in childhood T-ALL. The study is registered at http://www.clinicaltrials.gov; “Combination Chemotherapy Based on Risk of Relapse in Treating Young Patients With Acute Lymphoblastic Leukemia,” protocol identification #NCT00430118 for BFM and #NCT00613457 for AIEOP.
Collapse
|
39
|
Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood 2010; 115:3206-14. [PMID: 20154213 DOI: 10.1182/blood-2009-10-248146] [Citation(s) in RCA: 579] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The Associazione Italiana di Ematologia Oncologia Pediatrica and the Berlin-Frankfurt-Münster Acute Lymphoblastic Leukemia (AIEOP-BFM ALL 2000) study has for the first time introduced standardized quantitative assessment of minimal residual disease (MRD) based on immunoglobulin and T-cell receptor gene rearrangements as polymerase chain reaction targets (PCR-MRD), at 2 time points (TPs), to stratify patients in a large prospective study. Patients with precursor B (pB) ALL (n = 3184) were considered MRD standard risk (MRD-SR) if MRD was already negative at day 33 (analyzed by 2 markers, with a sensitivity of at least 10(-4)); MRD high risk (MRD-HR) if 10(-3) or more at day 78 and MRD intermediate risk (MRD-IR): others. MRD-SR patients were 42% (1348): 5-year event-free survival (EFS, standard error) is 92.3% (0.9). Fifty-two percent (1647) were MRD-IR: EFS 77.6% (1.3). Six percent of patients (189) were MRD-HR: EFS 50.1% (4.1; P < .001). PCR-MRD discriminated prognosis even on top of white blood cell count, age, early response to prednisone, and genotype. MRD response detected by sensitive quantitative PCR at 2 predefined TPs is highly predictive for relapse in childhood pB-ALL. The study is registered at http://clinicaltrials.gov: NCT00430118 for BFM and NCT00613457 for AIEOP.
Collapse
|
40
|
Pui CH, Pei D, Sandlund JT, Ribeiro RC, Rubnitz JE, Raimondi SC, Onciu M, Campana D, Kun LE, Jeha S, Cheng C, Howard SC, Metzger ML, Bhojwani D, Downing JR, Evans WE, Relling MV. Long-term results of St Jude Total Therapy Studies 11, 12, 13A, 13B, and 14 for childhood acute lymphoblastic leukemia. Leukemia 2010; 24:371-82. [PMID: 20010620 PMCID: PMC2820159 DOI: 10.1038/leu.2009.252] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 08/06/2009] [Indexed: 02/08/2023]
Abstract
We analyzed the long-term outcome of 1011 patients treated in five successive clinical trials (Total Therapy Studies 11, 12, 13A, 13B, and 14) between 1984 and 1999. The event-free survival improved significantly (P=0.003) from the first two trials conducted in the 1980s to the three more recent trials conducted in the 1990s. Approximately 75% of patients treated in the 1980s and 80% in the 1990s were cured. Early intensive triple intrathecal therapy, together with more effective systemic therapy, including consolidation and reinduction treatment (Studies 13A and 13B) as well as dexamethasone (Study 13B), resulted in a very low rate of isolated central nervous system (CNS) relapse rate (<2%), despite the reduced use of cranial irradiation. Factors consistently associated with treatment outcome were age, leukocyte count, immunophenotype, DNA index, and minimal residual disease level after remission induction treatment. Owing to concerns about therapy-related secondary myeloid leukemia and brain tumors, in our current trials we reserve the use of etoposide for patients with refractory or relapsed leukemia undergoing hematopoietic stem cell transplantation, and cranial irradiation for those with CNS relapse. The next main challenge is to further increase cure rates while improving quality of life for all patients.
Collapse
Affiliation(s)
- C H Pui
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mejstríková E, Fronková E, Kalina T, Omelka M, Batinić D, Dubravcić K, Pospísilová K, Vásková M, Luria D, Cheng SH, Ng M, Leung Y, Kappelmayer J, Kiss F, Izraeli S, Stark B, Schrappe M, Trka J, Starý J, Hrusák O. Detection of residual B precursor lymphoblastic leukemia by uniform gating flow cytometry. Pediatr Blood Cancer 2010; 54:62-70. [PMID: 19760767 DOI: 10.1002/pbc.22261] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Residual disease (RD) is an important prognostic factor in acute lymphoblastic leukemia (ALL). Flow cytometry (FC)-based RD detection is easy to perform, but interpretation requires expert analysis due to individual differences among patients. PROCEDURE We focused at the design of standardized and reproducible RD monitoring in ALL. RD was investigated by a uniform gating strategy, which was designed internationally and tested in one center by Ig/TCR rearrangements. RESULTS For each gate, positivity cutoff value was assigned using quantification of non-leukemic background. Comparing to Ig/TCR at 0.1% level, 80 of 103 specimens were correctly diagnosed by FC. The predictive value of FC RD at day 15 was then analyzed. In B lineage ALL, day 15 FC significantly correlated with Ig/TCR results at day 33 and/or week 12 (P < 0.01). No significant correlation was found in T lineage ALL. CONCLUSIONS Thus, FC with preset uniform gating at day 15 predicts PCR-detectable MRD in B precursor ALL. Presented data may be used to define new polychromatic cytometric diagnostics of MRD including semiautomatic assessment. Pediatr Blood Cancer 2010; 54:62-70. (c) 2009 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Ester Mejstríková
- CLIP-Childhood Leukemia Investigation Prague, Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Standardized MRD quantification in European ALL trials: proceedings of the Second International Symposium on MRD assessment in Kiel, Germany, 18-20 September 2008. Leukemia 2009; 24:521-35. [PMID: 20033054 DOI: 10.1038/leu.2009.268] [Citation(s) in RCA: 241] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Assessment of minimal residual disease (MRD) has acquired a prominent position in European treatment protocols for patients with acute lymphoblastic leukemia (ALL), on the basis of its high prognostic value for predicting outcome and the possibilities for implementation of MRD diagnostics in treatment stratification. Therefore, there is an increasing need for standardization of methodologies and harmonization of terminology. For this purpose, a panel of representatives of all major European study groups on childhood and adult ALL and of international experts on PCR- and flow cytometry-based MRD assessment was built in the context of the Second International Symposium on MRD assessment in Kiel, Germany, 18-20 September 2008. The panel summarized the current state of MRD diagnostics in ALL and developed recommendations on the minimal technical requirements that should be fulfilled before implementation of MRD diagnostics into clinical trials. Finally, a common terminology for a standard description of MRD response and monitoring was established defining the terms 'complete MRD response', 'MRD persistence' and 'MRD reappearance'. The proposed MRD terminology may allow a refined and standardized assessment of response to treatment in adult and childhood ALL, and provides a sound basis for the comparison of MRD results between different treatment protocols.
Collapse
|
43
|
Long-term results of Taiwan Pediatric Oncology Group studies 1997 and 2002 for childhood acute lymphoblastic leukemia. Leukemia 2009; 24:397-405. [PMID: 20016538 DOI: 10.1038/leu.2009.248] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The long-term outcome of 1390 children with acute lymphoblastic leukemia (ALL), treated in two successive clinical trials (Taiwan Pediatric Oncology Group (TPOG)-ALL-97 and TPOG-ALL-2002) between 1997 and 2007, is reported. The event-free survival improved significantly (P=0.0004) over this period, 69.3+/-1.9% in 1997-2001 to 77.4+/-1.7% in 2002-2007. A randomized trial in TPOG-97 testing L-asparaginase versus epidoxorubicin in combination with vincristine and prednisolone for remission induction in standard-risk (SR; low-risk) patients yielded similar outcomes. Another randomized trial, in TPOG-2002, showed that for SR patients, two reinduction courses did not improve long-term outcome over one course. Decreasing use of prophylactic cranial irradiation in the period 1997-2008 was not associated with increased rates of CNS relapse, prompting complete omission of prophylactic cranial irradiation from TPOG protocols, beginning in 2009. Decreased use of etoposide and cranial irradiation likely contributed to the low incidence of second cancers. High-risk B-lineage ALL, T-cell, CD10 negativity, t(9;22), infant, and higher leukocyte count were consistently adverse factors, whereas hyperdiploidy >50 was a consistently favorable factor. Higher leukocyte count and t(9;22) retained prognostic significance in both TPOG-97 and TPOG-2002 by multivariate analysis. Although long-term outcome in TPOG clinical trials is comparable with results being reported worldwide, the persistent strength of certain prognostic variables and the lower frequencies of favorable outcome predictors, such as ETV6-RUNX1 and hyperdiploidy >50, in Taiwanese children warrant renewed effort to cure a higher proportion of patients while preserving their quality of life.
Collapse
|
44
|
Basso G, Veltroni M, Valsecchi MG, Dworzak MN, Ratei R, Silvestri D, Benetello A, Buldini B, Maglia O, Masera G, Conter V, Arico M, Biondi A, Gaipa G. Risk of relapse of childhood acute lymphoblastic leukemia is predicted by flow cytometric measurement of residual disease on day 15 bone marrow. J Clin Oncol 2009; 27:5168-74. [PMID: 19805690 DOI: 10.1200/jco.2008.20.8934] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Speed of blast clearance is an indicator of outcome in childhood acute lymphoblastic leukemia (ALL). Availability of measurement of minimal residual disease (MRD) at an early time point with a reduced-cost method is of clinical relevance. In the AIEOP-BFM-ALL (Associazione Italiana Ematologia Oncologia Pediatrica and Berlin-Frankfurt-Münster Study Group) 2000 trial, patients were stratified by levels of polymerase chain reaction (PCR) MRD at day +33 and +78. AIEOP studied the prognostic impact of MRD measured by flow cytometry (FCM) at day 15 of induction therapy. PATIENTS AND METHODS Bone marrow samples from 830 Italian patients were collected on day 15, after 14 days of steroids, and one dose of intrathecal methotrexate, vincristine, daunorubicine, and asparaginase. Cells were analyzed by four-color FCM for detection of leukemia-associated immunophenotypes. RESULTS Three patient risk groups were identified by FCM: standard (< 0.1% blast cells; 42% of the total), intermediate (0.1 to < 10%; 47%), and high (> or = 10%; 11%). Their 5-year cumulative incidences of relapse were 7.5% (SE, 1.5), 17.5% (SE, 2.1), and 47.2% (SE, 5.9), respectively. In multivariate analysis, FCM was the most important prognostic factor among those available by day 15, with two-fold and five-fold increase in the risk of relapse compared with patients with less than 0.1%. PCR MRD, when added to the model, had significant prognostic impact; yet high levels of FCM MRD retained an independent ability to detect a significantly higher risk of relapse. CONCLUSION Measurement of FCM MRD in day 15 bone marrow was the most powerful early predictor of relapse, applicable to virtually all patients; it may complement PCR MRD-based stratification including later time points, thus allowing additional treatment tailoring.
Collapse
Affiliation(s)
- Giuseppe Basso
- Laboratorio di Oncoematologia Pediatrica, Department of Pediatrics, University, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Scrideli CA, Assumpção JG, Ganazza MA, Araújo M, Toledo SR, Lee MLM, Delbuono E, Petrilli AS, Queiróz RP, Biondi A, Viana MB, Yunes JA, Brandalise SR, Tone LG. A simplified minimal residual disease polymerase chain reaction method at early treatment points can stratify children with acute lymphoblastic leukemia into good and poor outcome groups. Haematologica 2009; 94:781-9. [PMID: 19483156 DOI: 10.3324/haematol.2008.003137] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Minimal residual disease is an important independent prognostic factor in childhood acute lymphoblastic leukemia. The classical detection methods such as multiparameter flow cytometry and real-time quantitative polymerase chain reaction analysis are expensive, time-consuming and complex, and require considerable technical expertise. DESIGN AND METHODS We analyzed 229 consecutive children with acute lymphoblastic leukemia treated according to the GBTLI-99 protocol at three different Brazilian centers. Minimal residual disease was analyzed in bone marrow samples at diagnosis and on days 14 and 28 by conventional homo/heteroduplex polymerase chain reaction using a simplified approach with consensus primers for IG and TCR gene rearrangements. RESULTS At least one marker was detected by polymerase chain reaction in 96.4% of the patients. By combining the minimal residual disease results obtained on days 14 and 28, three different prognostic groups were identified: minimal residual disease negative on days 14 and 28, positive on day 14/negative on day 28, and positive on both. Five-year event-free survival rates were 85%, 75.6%, and 27.8%, respectively (p<0.0001). The same pattern of stratification held true for the group of intensively treated children. When analyzed in other subgroups of patients such as those at standard and high risk at diagnosis, those with positive B-derived CD10, patients positive for the TEL/AML1 transcript, and patients in morphological remission on a day 28 marrow, the event-free survival rate was found to be significantly lower in patients with positive minimal residual disease on day 28. Multivariate analysis demonstrated that the detection of minimal residual disease on day 28 is the most significant prognostic factor. CONCLUSIONS This simplified strategy for detection of minimal residual disease was feasible, reproducible, cheaper and simpler when compared with other methods, and allowed powerful discrimination between children with acute lymphoblastic leukemia with a good and poor outcome.
Collapse
Affiliation(s)
- Carlos A Scrideli
- Department of Pediatrics, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sutton R, Venn NC, Tolisano J, Bahar AY, Giles JE, Ashton LJ, Teague L, Rigutto G, Waters K, Marshall GM, Haber M, Norris MD. Clinical significance of minimal residual disease at day 15 and at the end of therapy in childhood acute lymphoblastic leukaemia. Br J Haematol 2009; 146:292-9. [PMID: 19500099 DOI: 10.1111/j.1365-2141.2009.07744.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Detection of minimal residual disease (MRD) after induction and consolidation therapy is highly predictive of outcome for childhood acute lymphoblastic leukaemia (ALL) and is used to identify patients at high risk of relapse in several current clinical trials. To evaluate the prognostic significance of MRD at other treatment phases, MRD was measured by real-time quantitative polymerase chain reaction on a selected group of 108 patients enrolled on the Australian and New Zealand Children's Cancer Study Group Study VII including 36 patients with a bone marrow or central nervous system relapse and 72 matched patients in first remission. MRD was prognostic of outcome at all five treatment phases tested: at day 15 (MRD > or = 5 x 10(-2), log rank P < 0.0001), day 35 (> or =1 x 10(-2), P = 0.0001), 4 months (> or =5 x 10(-4), P < 0.0001), 12 months (MRD > or = 1 x 10(-4), P = 0.006) and 24 months (MRD > or = 1 x 10(-4), P < 0.0001). Day 15 was the best early MRD time-point to differentiate between patients with high, intermediate and low risk of relapse. MRD testing at 12 and particularly at 24 months, detected molecular relapses in some patients up to 6 months before clinical relapse. This raised the question of whether a strategy of late monitoring and salvage therapy will improve outcome.
Collapse
Affiliation(s)
- Rosemary Sutton
- Children's Cancer Institute Australia for Medical Research, University of NSW, Sydney, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
High-resolution genomic profiling of childhood T-ALL reveals frequent copy-number alterations affecting the TGF-beta and PI3K-AKT pathways and deletions at 6q15-16.1 as a genomic marker for unfavorable early treatment response. Blood 2009; 114:1053-62. [PMID: 19406988 DOI: 10.1182/blood-2008-10-186536] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Precursor T-cell acute lymphoblastic leukemia (T-ALL) in children represents a clinical challenge, because relapses are usually fatal. It is thus necessary to identify high-risk patients as early as possible to effectively individualize treatment. We aimed to define novel molecular risk markers in T-ALL and performed array-based comparative genomic hybridization (array-CGH) and expression analyses in 73 patients. We show that DNA copy-number changes are common in T-ALL and affect 70 of 73 (96%) patients. Notably, genomic imbalances predicted to down-regulate the TGF-beta or up-regulate the PI3K-AKT pathways are identified in 25 of 73 (34%) and 21 of 73 (29%) patients, suggesting that these pathways play key roles in T-ALL leukemogenesis. Furthermore, we identified a deletion at 6q15-16.1 in 9 of 73 (12%) of the patients, which predicts poor early treatment response. This deletion includes the CASP8AP2 gene, whose expression is shown to be down-regulated. The interaction of CASP8AP2 with CASP8 plays a crucial role in apoptotic regulation, suggesting a functional link between the clinical effect of the deletion and the molecular mode of action. The data presented here implicate the TGF-beta and PI3K-AKT pathways in T-ALL leukemogenesis and identify a subgroup of patients with CASP8AP2 deletions and poor early treatment response.
Collapse
|