1
|
Deng S, Ou J, Chen J, Huang Z, Cai Z, Xu X, Tang B, Ding C, Li J, Lin R, Wang Z, Zhang T, Liu Q, Zhou H. Refining Risk Stratification for B-cell Precursor Adult Acute Lymphoblastic Leukemia Treated With a Pediatric-inspired Regimen by Combining IKZF1 Deletion and Minimal Residual Disease. Transplant Cell Ther 2025; 31:242-252. [PMID: 39798801 DOI: 10.1016/j.jtct.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/27/2024] [Accepted: 01/04/2025] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Minimal residual disease (MRD) is the most important prognostic factor for B-cell acute lymphoblastic leukemia (B-ALL) however nearly 20-30% of patients relapsed even when they achieved negative MRD, how to identify these patients is less addressed. In this study, we aimed to reassess the prognostic significance of MRD and IKZF1 in adult B-ALL patients receiving pediatric chemotherapy regimens. METHODOLOGY A total of 202 newly diagnosed adult patients with B-ALL treated at Nanfang Hospital between January 2016 and September 2020 were enrolled in the population-based protocol of the PDT-ALL-2016 trial (NCT03564470), a GRAALL-backbone, peg-aspargase-intensified, antimetabolite-based pediatric-inspired regimen therapy. The validation dataset COG-P9906, which includes complete gene expression profiles and clinical data for 190 samples, is accessible via the NCBI Gene Expression Omnibus (GEO) at the following link: (https://www.ncbi.nlm.nih.gov/geo/), under the accession code GSE11877. MAIN FINDINGS B-ALL patients were redefined as standard (MRD-negative and IKZF1wild-type), intermediate (MRD-positive or IKZF1 deletion), and high-risk (MRD-positive and IKZF1 deletion) groups by combining IKZF1 deletion status and MRD. In the PDT-ALL-2016 cohort, patients in the high- and intermediate-risk groups who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) exhibited significantly improved 5-year overall survival (OS), event-free survival (EFS), and lower cumulative incidence of relapse (CIR) compared to those who received chemotherapy alone. In the PDT-ALL-2016 cohort, no significant advantage was observed in the 5-year OS, EFS, and CIR of patients in the standard-risk group who received allo-HSCT compared to those who received chemotherapy. DISCUSSION Traditional risk factors, incorporating clinical and cytogenetic features, have been previously evaluated to stratify risks and guide treatment decisions. However, the prognostic strength of this stratified system is limited by the pediatric-inspired protocol background, making it difficult to identify patients with a high risk of relapse. Therefore, in the era of pediatric-inspired protocols, it is imperative to reassess traditional risk factors to identify patients at high risk of recurrence and mortality.In this study, we retrospectively evaluated the combination of MRD and IKZF1 to develop an efficient risk stratification tool for adult patients with B-ALL in the pediatric-inspired chemotherapy era. Moreover, allo-HSCT had distinct efficacy at different risk levels, which means that the decision to perform allo-HSCT may be well guided by this risk classification scheme. CONCLUSION In conclusion, based on our cohort study and validation cohort, we demonstrated that the combination of MRD and IKZF1 deletion allows for better risk stratification of adults with B-ALL and that allo-HSCT mitigates the poor prognosis of MRD+ and/or IKZF1del subgroups.
Collapse
Affiliation(s)
- Shiyu Deng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiawang Ou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zicong Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zihong Cai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuli Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingqing Tang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chenhao Ding
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ren Lin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhixiang Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Østergaard A, Boer JM, van Leeuwen FN, Pieters R, Den Boer ML. IKZF1 in acute lymphoblastic leukemia: the rise before the fall? Leuk Lymphoma 2024; 65:2077-2087. [PMID: 39210599 DOI: 10.1080/10428194.2024.2396046] [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: 04/09/2024] [Revised: 07/23/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy in children and adolescents and in recent decades, the survival rates have risen to >90% in children largely due the introduction of risk adapted therapy. Therefore, knowledge of factors influencing risk of relapse is important. The transcription factor IKAROS is a regulator of lymphocyte development and alterations of its coding gene, IKZF1, are frequent in ALL and are associated with higher relapse risk. This concise review will discuss the normal function of IKAROS together with the effect of gene alterations in ALL such as relieved energy restriction and altered response to anti-leukemic drugs. Besides the biology, the clinical impact of gene alterations in the different subtypes of ALL will be discussed. Finally, possibilities for treating ALL with IKZF1 alterations will be considered including novel therapies like cell signaling inhibitors and immunotherapy.
Collapse
Affiliation(s)
- Anna Østergaard
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Judith M Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | |
Collapse
|
3
|
de Albuquerque A, Lopes BA, Fernandes RA, Gimba ERP, Emerenciano M. IKZF1 and BTG1 silencing reduces glucocorticoid response in B-cell precursor acute leukemia cell line. Hematol Transfus Cell Ther 2024; 46 Suppl 6:S163-S170. [PMID: 39095315 PMCID: PMC11726076 DOI: 10.1016/j.htct.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 12/26/2023] [Accepted: 05/07/2024] [Indexed: 08/04/2024] Open
Abstract
INTRODUCTION Secondary genetic alterations, which contribute to the dysregulation of cell cycle progression and lymphoid specialization, are frequently observed in B-cell precursor acute lymphoblastic leukemia (B-ALL). As IKZF1 and BTG1 deletions are associated with a worse outcome in B-ALL, this study aimed to address whether they synergistically promote glucocorticoid resistance. METHODS Small interfering RNA was used to downregulate either IKZF1, or BTG1, or both genes in the 207 B-ALL cell line. Cell viability was investigated by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and trypan blue exclusion assays. The expression levels of IKZF1, BTG1 and glucocorticoid-responsive genes (DUSP1, SGK1, FBXW7 and NR3C1) were evaluated by real time quantitative real time polymerase chain reaction (PCR). RESULTS Isolated silencing of BTG1, IKZF1, or both genes in combination under dexamethasone treatment increased cell viability by 24%, 40% and 84%, respectively. Although BTG1 silencing did not alter the expression of glucocorticoid-responsive genes, IKZF1 knockdown decreased the transcript levels of DUSP1 (2.6-fold), SGK1 (1.8-fold), FBXW7 (2.2-fold) and NR3C1 (1.7-fold). The expression of glucocorticoid-responsive genes reached even lower levels (reducing 2.4-4 fold) when IKZF1 and BTG1 silencing occurred in combination. CONCLUSIONS IKZF1 silencing impairs the transcription of glucocorticoid-responsive genes; this effect is enhanced by concomitant loss of BTG1. These results demonstrate the molecular mechanism by which the combination of both genetic deletions might contribute to higher relapse rates in B-ALL.
Collapse
Affiliation(s)
- Amanda de Albuquerque
- Division of Clinical Research and Technological Development, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil
| | - Bruno A Lopes
- Division of Clinical Research and Technological Development, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil; Genetics of Acute Leukemia Laboratory, Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil
| | - Renan Amphilophio Fernandes
- Pharmacology and Medicinal Chemistry Program, Institute of Biological Sciences, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Etel Rodrigues Pereira Gimba
- Department of Natural Sciences (RCN), Institute of Humanities and Health (IHS), Universidade Federal Fluminense (UFF), Rio de Janeiro, Brazil; Hematology-Molecular Oncology Program, Research Coordination, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Mariana Emerenciano
- Division of Clinical Research and Technological Development, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil; Genetics of Acute Leukemia Laboratory, Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
4
|
Pan L, Chen Y, Weng K, Guo B, Zhuang S, Huang S, Lian Z, Wang X, Li N, Zheng Y. Prognostic significance and treatment strategies for IKZF1 deletion in pediatric B-cell precursor acute lymphoblastic leukemia. BMC Cancer 2024; 24:1070. [PMID: 39210321 PMCID: PMC11363382 DOI: 10.1186/s12885-024-12828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The predictive importance of IKZF1del in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL) has shown variability across different studies. Thus, the optimal treatment approach for children with IKZF1del BCP-ALL remains contentious, with the ongoing debate surrounding the use of IKZF1del-based high-risk stratification versus a minimal residual disease (MRD)-guided protocol. METHODS IKZF1 status was reliably determined in 804 patients using multiplex ligation-dependent probe amplification (MLPA) data obtained from four hospitals in Fujian, a province of China. In the Chinese Children Leukemia Group (CCLG)-ALL 2008 cohort, IKZF1 status was included in the risk assignment, with all IKZF1del patients receiving a high-risk regimen. Conversely, in the Chinese Children's Cancer Group (CCCG)-ALL 2015 cohort, IKZF1del was not incorporated into the risk assignment, and patients were treated based on an MRD-guided risk stratification protocol. RESULTS IKZF1del was found in 86 patients (86/804, 10.7%) overall and in 30 (30/46, 65.2%) BCR::ABL1-positive patients. Overall, IKZF1del was a poor prognostic predictor for patients, though the significance diminished upon age adjustment, white blood cell (WBC) count at diagnosis, treatment group, and MRD status. In the CCLG-ALL 2008 cohort, IKZF1del conferred a notably lower 5-year overall survival (OS) and event-free survival (EFS) and a significantly higher 5-year cumulative incidence of relapse (CIR) than IKZF1wt. In the CCLG-ALL 2015 cohort, IKZF1del conferred a lower 5-year OS and EFS and a higher 5-year CIR than IKZF1wt, but the differences were insignificant. The IKZF1del patients treated with higher intensity chemotherapy (CCLG-ALL 2008 high-risk regimen) had a markedly lower 5-year OS and EFS compared with those treated with the MRD-guided protocol (CCCG-ALL 2015 protocol). Furthermore, patients treated with the CCLG-ALL 2008 high-risk regimen experienced a higher frequency of serious adverse events (SAEs), especially infection-related SAEs, compared with those treated with the CCCG-ALL 2015 MRD-guided protocol. CONCLUSIONS The prognostic effect of IKZF1del may vary in different protocols. Compared with higher intensity chemotherapy, the MRD-guided protocol may be a more effective approach to treating BCP-ALL with IKZF1del in children.
Collapse
Affiliation(s)
- Lili Pan
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yiqiao Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Kaizhi Weng
- Department of Pediatric Hematology, Rheumatology and Nephrology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Biyun Guo
- Department of Pediatrics, the First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Shuquan Zhuang
- Department of Pediatrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Shuxian Huang
- Department of Pediatric Hematology, Rheumatology and Nephrology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Zhulan Lian
- Department of Pediatrics, the First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiaofang Wang
- Department of Pediatrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Nainong Li
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Yongzhi Zheng
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
5
|
Feng L, Zhang H, Liu T. Multifaceted roles of IKZF1 gene, perspectives from bench to bedside. Front Oncol 2024; 14:1383419. [PMID: 38978740 PMCID: PMC11228169 DOI: 10.3389/fonc.2024.1383419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/10/2024] [Indexed: 07/10/2024] Open
Abstract
The IKZF1 gene encodes a transcription factor that belongs to the family of zinc-finger DNA-binding proteins associated with chromatin remodeling. The protein product, IKAROS, had been proved to regulate lymphopoiesis. Subsequent mouse model studies have further confirmed its regulating role in lymphopoiesis as well as in hematopoiesis; besides, it associates with immune function, certain immune disorders like common variable immunodeficiency and dysgammaglobulinemia have been proved to be associated with germline IKZF1 mutations. Dysfunction of IKAROS also bears paramount significance in leukemic transformation and alterations of IKZF1 gene predicts a poor prognosis in hematological malignancies. As an independent prognostic marker, IKZF1 has been incorporated in the risk stratification of BCP-ALL and stratification-guided therapy has also been generated. In this review, we provide a concise and comprehensive overview on the multifaceted roles of IKZF1 gene.
Collapse
Affiliation(s)
| | | | - Ting Liu
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Garcia-Solorio J, Núñez-Enriquez JC, Jiménez-Olivares M, Flores-Lujano J, Flores-Espino F, Molina-Garay C, Cervera A, Casique-Aguirre D, Peñaloza-Gonzalez JG, Baños-Lara MDR, García-Soto Á, Galván-Díaz CA, Olaya-Vargas A, Aguilar HF, Mata-Rocha M, Garrido-Hernández MÁ, Solís-Poblano JC, Luna-Silva NC, Cano-Cuapio LS, Aristil-Chery PM, Herrera-Quezada F, Carrillo-Sanchez K, Muñoz-Rivas A, Flores-Lagunes LL, Mendoza-Caamal EC, Villegas-Torres BE, González-Osnaya V, Jiménez-Hernández E, Torres-Nava JR, Martín-Trejo JA, Gutiérrez-Rivera MDL, Espinosa-Elizondo RM, Merino-Pasaye LE, Pérez-Saldívar ML, Jiménez-Morales S, Curiel-Quesada E, Rosas-Vargas H, Mejía-Arangure JM, Alaez-Verson C. IKZF1plus is a frequent biomarker of adverse prognosis in Mexican pediatric patients with B-acute lymphoblastic leukemia. Front Oncol 2024; 14:1337954. [PMID: 38634053 PMCID: PMC11022689 DOI: 10.3389/fonc.2024.1337954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/29/2024] [Indexed: 04/19/2024] Open
Abstract
Background Recurrent genetic alterations contributing to leukemogenesis have been identified in pediatric B-cell Acute Lymphoblastic Leukemia (B-ALL), and some are useful for refining classification, prognosis, and treatment selection. IKZF1plus is a complex biomarker associated with a poor prognosis. It is characterized by IKZF1 deletion coexisting with PAX5, CDKN2A/2B, or PAR1 region deletions. The mutational spectrum and clinical impact of these alterations have scarcely been explored in Mexican pediatric patients with B-ALL. Here, we report the frequency of the IKZF1plus profile and the mutational spectrum of IKZF1, PAX5, CDKN2A/2B, and ERG genes and evaluate their impact on overall survival (OS) in a group of patients with B-ALL. Methods A total of 206 pediatric patients with de novo B-ALL were included. DNA was obtained from bone marrow samples at diagnosis before treatment initiation. A custom-designed next-generation sequencing panel was used for mutational analysis. Kaplan-Meier analysis was used for OS estimation. Results We identified the IKZF1plus profile in 21.8% of patients, which was higher than that previously reported in other studies. A significantly older age (p=0.04), a trend toward high-risk stratification (p=0.06), and a decrease in 5-year Overall Survival (OS) (p=0.009) were observed, although heterogeneous treatment protocols in our cohort would have impacted OS. A mutation frequency higher than that reported was found for IKZF1 (35.9%) and CDKN2A/2B (35.9%) but lower for PAX5 (26.6%). IKZF1MUT group was older at diagnosis (p=0.0002), and most of them were classified as high-risk (73.8%, p=0.02), while patients with CDKN2A/2BMUT had a higher leukocyte count (p=0.01) and a tendency toward a higher percentage of blasts (98.6%, >50% blasts, p=0.05) than the non-mutated patients. A decrease in OS was found in IKZF1MUT and CDKN2A/2BMUT patients, but the significance was lost after IKZF1plus was removed. Discussion Our findings demonstrated that Mexican patients with B-ALL have a higher prevalence of genetic markers associated with poor outcomes. Incorporating genomic methodologies into the diagnostic process, a significant unmet need in low- and mid-income countries, will allow a comprehensive identification of relevant alterations, improving disease classification, treatment selection, and the general outcome.
Collapse
Affiliation(s)
- Joaquin Garcia-Solorio
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Juan Carlos Núñez-Enriquez
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Medica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Marco Jiménez-Olivares
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Janet Flores-Lujano
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Medica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Fernanda Flores-Espino
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Carolina Molina-Garay
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Alejandra Cervera
- Subdirección de Genómica Poblacional, Instituto Nacional de Medicina Genomica (INMEGEN), Mexico City, Mexico
| | - Diana Casique-Aguirre
- Laboratorio de Citómica del Cáncer Infantil, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Delegación Puebla, Puebla, Mexico
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City, Mexico
| | | | - Ma. Del Rocío Baños-Lara
- Centro de Investigación Oncológica Una Nueva Esperanza, Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico
| | - Ángel García-Soto
- Hospital General Centro Médico La Raza, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | - Alberto Olaya-Vargas
- Departamento de Oncologia, Instituto Nacional de Pediatría (INP), Mexico City, Mexico
| | - Hilario Flores Aguilar
- Departamento de Inmunogenetica, Instituto de Diagnostico y Referencia Epidemiologicos (InDRE), Mexico City, Mexico
| | - Minerva Mata-Rocha
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | - Juan Carlos Solís-Poblano
- Servicio de Oncohematología Pediátrica, Instituto Mexicano del Seguro (IMSS) Unidad Médica de Alta Especialidad (UMAE) Centro Médico Nacional (CMN) Hospital de Especialidades Dr. Manuel Ávila Camacho, Puebla, Mexico
| | - Nuria Citlalli Luna-Silva
- Servicio de Hemato-Oncología Pediátrica, Hospital de la Niñez Oaxaqueña "Dr. Guillermo Zárate Mijangos", Secretaria de Salud y Servicios de Salud Oaxaca (SSO), Oaxaca, Mexico
| | | | - Pierre Mitchel Aristil-Chery
- Instituto de Seguridad y Servicios Sociales de los Trabajadores al Servicio de los Poderes del Estado (ISSSTE) de Puebla, Departamento de Enseñanza e Investigació, Puebla, Mexico
| | - Fernando Herrera-Quezada
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Medica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Karol Carrillo-Sanchez
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Anallely Muñoz-Rivas
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | | | | | | | - Vincent González-Osnaya
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Elva Jiménez-Hernández
- Servicio de Oncología, Hospital Pediátrico Moctezuma, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - José Refugio Torres-Nava
- Servicio de Oncología, Hospital Pediátrico Moctezuma, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Jorge Alfonso Martín-Trejo
- Servicio de Hematología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Unidad Médica de Alta Especialidad (UMAE) Hospital de Pediatría “Dr. Silvestre Frenk Freund”, Mexico City, Mexico
| | - María de Lourdes Gutiérrez-Rivera
- Servicio de Oncología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Unidad Médica de Alta Especialidad (UMAE) Hospital de Pediatría “Dr. Silvestre Frenk Freund”, Mexico City, Mexico
| | | | - Laura Elizabeth Merino-Pasaye
- Servicio de Hematología Pediátrica, Centro Médico Nacional (CMN) “20 de Noviembre”, Instituto de Seguridad Social al Servicio de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - María Luisa Pérez-Saldívar
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Medica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Silvia Jiménez-Morales
- Laboratorio de Medicina de Precisión, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Everardo Curiel-Quesada
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional (IPN), Mexico City, Mexico
| | - Haydeé Rosas-Vargas
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Juan Manuel Mejía-Arangure
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Carmen Alaez-Verson
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
7
|
Akin-Bali DF, Doganay Erdogan B, Aslar Oner D, Mahmud A, Tasdelen S, Kurekci E, Akar N, Ozdag Sevgili H. Genetic Profiling of Pediatric Patients with B-Cell Precursor Acute Lymphoblastic Leukemia. J Pediatr Genet 2023; 12:288-300. [PMID: 38162155 PMCID: PMC10756719 DOI: 10.1055/s-0041-1742246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is a heterogeneous leukemia subgroup. It has multiple sub-types that are likely to be classified by prognostic factors. Following a systematic literature review, this study analyzed the genes correlated with BCP-ALL prognosis ( IKZF1, PAX5, EBF1, CREBBP, CRLF2, JAK2, ERG, CXCR4, ZAP70, VLA4, NF1, NR3C1, RB1, TSLP, ZNRF1, and FOXO3A) , specifically their nucleotide variations and expression profiles in pediatric BCP-ALL samples. The study included 45 pediatric BCP-ALL patients with no cytogenetic anomaly and a control group of 10 children. The selected genes' hot-spot regions were sequenced using next-generation sequencing, while Polymorphism Phenotyping v2 and Supplemental Nutrition Assistance Program were used to identify pathogenic mutations. The expression analysis was performed using quantitative real-time polymerase chain reaction. The mutation analysis detected 328 variants (28 insertions, 47 indels, 74 nucleotide variants, 75 duplications, and 104 deletions). The most and least frequently mutated genes were IKZF1 and CREBBP , respectively. There were statistically significant differences between patients and controls for mutation distribution in eight genes ( ERG, CRLF2, CREBBP, TSLP, JAK2, ZAP70, FOXO3A, and NR3C1 ). The expression analysis revealed that JAK and ERG were significantly overexpressed in patients compared with controls (respectively, p = 0.004 and p = 0.003). This study combined genes and pathways previously analyzed in pediatric BCP-ALL into one dataset for a comprehensive analysis from the same samples to unravel candidate prognostic biomarkers. Novel mutations were identified in all of the studied genes.
Collapse
Affiliation(s)
- Dilara Fatma Akin-Bali
- Department of Medical Biology, Faculty of Medicine, Niğde Ömer Halisdemir University, Niğde, Turkey
| | - Beyza Doganay Erdogan
- Department of Biostatistic, Faculty of Medicine, Biostatistics, Ankara University, Ankara, Turkey
| | - Deniz Aslar Oner
- Atatürk Vocational School of Health Services, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Akkan Mahmud
- LÖSANTE Children's and Adult Hospital, Ankara, Turkey
| | | | - Emin Kurekci
- LÖSANTE Children's and Adult Hospital, Ankara, Turkey
| | - Nejat Akar
- Department of Pediatrics, TOBB-ETU Hospital, Ankara, Turkey
| | | |
Collapse
|
8
|
Kimura H, Onozawa M, Yoshida S, Miyashita N, Yokoyama S, Matsukawa T, Hirabayashi S, Goto H, Endo T, Oguri S, Fujisawa S, Mori A, Kondo T, Hidaka D, Okada K, Ota S, Kakinoki Y, Tsutsumi Y, Yamamoto S, Miyagishima T, Hashiguchi J, Nagashima T, Ibata M, Wakasa K, Haseyama Y, Fujimoto K, Ishihara T, Sakai H, Teshima T. Dominant-negative type of IKZF1 deletion showed a favorable prognosis in adult B-cell acute lymphoblastic leukemia. Ann Hematol 2023; 102:3103-3113. [PMID: 37597110 DOI: 10.1007/s00277-023-05405-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/06/2023] [Indexed: 08/21/2023]
Abstract
IKZF1 deletion is a recurrent genomic alteration in B-cell acute lymphoblastic leukemia (B-ALL) and is divided into dominant-negative (DN) and loss of function (LOF) deletions. The prognostic impact of each deletion has not been fully elucidated. We retrospectively analyzed 117 patients with adult B-ALL including 60 patients with BCR::ABL1-positive B-ALL and 57 patients with BCR::ABL1-negative B-ALL by the fluorescence in situ hybridization (FISH) method for IKZF1 deletion and multiplex PCR for the 4 most common IKZF1 deletions (∆4-7, ∆2-7, ∆2-8, and ∆4-8). Samples, in which IKZF1 deletion was detected by FISH but a specific type of deletion was not identified by the PCR, were categorized as "other." Patients were classified into a DN group that had at least 1 allele of ∆4-7 (n = 23), LOF and other group (n = 40), and wildtype group (n = 54). DN type IKZF1 deletions were found in 33.3% of BCR::ABL1-positive cases and 5.2% of BCR::ABL1-negative cases. LOF and other type IKZF1 deletions were found in 43.4% of BCR::ABL1-positive cases and 24.6% of BCR::ABL1-negative cases. Patients with the DN group showed significantly higher overall survival (OS) than that of the LOF and other and WT groups (P = 0.011). Multivariate analysis including age, WBC counts, complex karyotype, and DN type IKZF1 deletion showed that the DN type of IKZF1 deletion (HR = 0.22, P = 0.013) had a positive impact and age ≥ 65 (HR = 1.92, P = 0.029) had a negative impact on OS. The prognostic impact of IKZF1 deletion depends on the type of deletion and DN type of IKZF1 deletion showed better prognosis in adult B-ALL patients.Clinical trial registration This study was part of a prospective observational study (Hokkaido Leukemia Net, UMIN000048611). It was conducted in compliance with ethical principles based on the Helsinki Declaration and was approved by the institutional review board of Hokkaido University Hospital (#015-0344).
Collapse
Affiliation(s)
- Hiroyuki Kimura
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Masahiro Onozawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan.
| | - Shota Yoshida
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Naoki Miyashita
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Shota Yokoyama
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Toshihiro Matsukawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | | | - Hideki Goto
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Tomoyuki Endo
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Satoshi Oguri
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Shinichi Fujisawa
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Akio Mori
- Blood Disorders Center, Aiiku Hospital, Sapporo, Japan
| | - Takeshi Kondo
- Blood Disorders Center, Aiiku Hospital, Sapporo, Japan
| | - Daisuke Hidaka
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Kohei Okada
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Shuichi Ota
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | | | - Yutaka Tsutsumi
- Department of Hematology, Hakodate Municipal Hospital, Hakodate, Japan
| | - Satoshi Yamamoto
- Department of Hematology, Sapporo City General Hospital, Sapporo, Japan
| | | | - Junichi Hashiguchi
- Department of Internal Medicine, Kitami Red Cross Hospital, Kitami, Japan
| | - Takahiro Nagashima
- Department of Internal Medicine, Kitami Red Cross Hospital, Kitami, Japan
| | - Makoto Ibata
- Department of Hematology, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Kentaro Wakasa
- Department of Hematology, Obihiro Kosei Hospital, Obihiro, Japan
| | | | - Katsuya Fujimoto
- Department of Hematology, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | | | - Hajime Sakai
- Department of Hematology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
9
|
Pieters R, de Groot-Kruseman H, Fiocco M, Verwer F, Van Overveld M, Sonneveld E, van der Velden V, Beverloo HB, Bierings M, Dors N, de Haas V, Hoogerbrugge P, Van der Sluis I, Tissing W, Veening M, Boer J, Den Boer M. Improved Outcome for ALL by Prolonging Therapy for IKZF1 Deletion and Decreasing Therapy for Other Risk Groups. J Clin Oncol 2023; 41:4130-4142. [PMID: 37459571 DOI: 10.1200/jco.22.02705] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/30/2023] [Accepted: 05/12/2023] [Indexed: 07/20/2023] Open
Abstract
PURPOSE The ALL10 protocol improved outcomes for children with ALL by stratifying and adapting therapy into three minimal residual disease-defined risk groups: standard risk, medium risk (MR), and high risk. IKZF1-deleted (IKZF1del) ALL in the largest MR group still showed poor outcome, in line with protocols worldwide, accounting for a high number of overall relapses. ALL10 showed high toxicity in Down syndrome (DS) and excellent outcome in ETV6::RUNX1 ALL. Poor prednisone responders (PPRs) were treated as high risk in ALL10. In ALL11, we prolonged therapy for IKZF1del from 2 to 3 years. We reduced therapy for DS by omitting anthracyclines completely, for ETV6::RUNX1 in intensification, and for PPR by treatment as MR. METHODS Eight hundred nineteen patients with ALL (age, 1-18 years) were enrolled on ALL11 and stratified as in ALL10. Results were compared with those in ALL10. RESULTS The five-year overall survival (OS), event-free survival (EFS), cumulative risk of relapse (CIR), and death in complete remission on ALL11 were 94.2% (SE, 0.9%), 89.0% (1.2), 8.2% (1.1), and 2.3% (0.6), respectively. Prolonged maintenance for IKZF1del MR improved 5-year CIR by 2.2-fold (10.8% v 23.4%; P = .035) and EFS (87.1% v 72.3%; P = .019). Landmark analysis at 2 years from diagnosis showed a 2.9-fold reduction of CIR (25.6%-8.8%; P = .008) and EFS improvement (74.4%-91.2%; P = .007). Reduced therapy did not abrogate 5-year outcome for ETV6::RUNX1 (EFS, 98.3%; OS, 99.4%), DS (EFS, 87.0%; OS, 87.0%), and PPR (EFS, 81.1%; OS, 94.9%). CONCLUSION Children with IKZF1del ALL seem to benefit from prolonged maintenance therapy. Chemotherapy was successfully reduced for patients with ETV6::RUNX1, DS, and PPR ALL. It has to be noted that these results were obtained in a nonrandomized study using a historical control group.
Collapse
Affiliation(s)
- Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, the Netherlands
| | - Hester de Groot-Kruseman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, the Netherlands
| | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, the Netherlands
- Department of Biomedical Data Science, Section Medical Statistics, Leiden University Medical Center, Leiden, the Netherlands
- Mathematical Institute, Leiden University, Leiden, the Netherlands
| | - Femke Verwer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, the Netherlands
| | | | - Edwin Sonneveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, the Netherlands
| | | | - H Berna Beverloo
- Department of Clinical Genetics, Erasmus MC, Rotterdam, the Netherlands
| | - Marc Bierings
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, the Netherlands
| | - Natasja Dors
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Valérie de Haas
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, the Netherlands
| | | | | | - Wim Tissing
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Margreet Veening
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Judith Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Monique Den Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
10
|
Bedics G, Egyed B, Kotmayer L, Benard-Slagter A, de Groot K, Bekő A, Hegyi LL, Bátai B, Krizsán S, Kriván G, Erdélyi DJ, Müller J, Haltrich I, Kajtár B, Pajor L, Vojcek Á, Ottóffy G, Ujfalusi A, Szegedi I, Tiszlavicz LG, Bartyik K, Csanádi K, Péter G, Simon R, Hauser P, Kelemen Á, Sebestyén E, Jakab Z, Matolcsy A, Kiss C, Kovács G, Savola S, Bödör C, Alpár D. PersonALL: a genetic scoring guide for personalized risk assessment in pediatric B-cell precursor acute lymphoblastic leukemia. Br J Cancer 2023; 129:455-465. [PMID: 37340093 PMCID: PMC10403542 DOI: 10.1038/s41416-023-02309-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 05/08/2023] [Accepted: 06/05/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Recurrent genetic lesions provide basis for risk assessment in pediatric acute lymphoblastic leukemia (ALL). However, current prognostic classifiers rely on a limited number of predefined sets of alterations. METHODS Disease-relevant copy number aberrations (CNAs) were screened genome-wide in 260 children with B-cell precursor ALL. Results were integrated with cytogenetic data to improve risk assessment. RESULTS CNAs were detected in 93.8% (n = 244) of the patients. First, cytogenetic profiles were combined with IKZF1 status (IKZF1normal, IKZF1del and IKZF1plus) and three prognostic subgroups were distinguished with significantly different 5-year event-free survival (EFS) rates, IKAROS-low (n = 215): 86.3%, IKAROS-medium (n = 27): 57.4% and IKAROS-high (n = 18): 37.5%. Second, contribution of genetic aberrations to the clinical outcome was assessed and an aberration-specific score was assigned to each prognostically relevant alteration. By aggregating the scores of aberrations emerging in individual patients, personalized cumulative values were calculated and used for defining four prognostic subgroups with distinct clinical outcomes. Two favorable subgroups included 60% of patients (n = 157) with a 5-year EFS of 96.3% (excellent risk, n = 105) and 87.2% (good risk, n = 52), respectively; while 40% of patients (n = 103) showed high (n = 74) or ultra-poor (n = 29) risk profile (5-year EFS: 67.4% and 39.0%, respectively). CONCLUSIONS PersonALL, our conceptually novel prognostic classifier considers all combinations of co-segregating genetic alterations, providing a highly personalized patient stratification.
Collapse
Affiliation(s)
- Gábor Bedics
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Bálint Egyed
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
- Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Lili Kotmayer
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | | | - Anna Bekő
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Lajos László Hegyi
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Bence Bátai
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Szilvia Krizsán
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gergely Kriván
- Central Hospital of Southern Pest - National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Dániel J Erdélyi
- Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Judit Müller
- Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Irén Haltrich
- Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Béla Kajtár
- Department of Pathology, University of Pécs Medical School, Pécs, Hungary
| | - László Pajor
- Department of Pathology, University of Pécs Medical School, Pécs, Hungary
| | - Ágnes Vojcek
- Department of Pediatrics, University of Pécs Medical School, Pécs, Hungary
| | - Gábor Ottóffy
- Department of Pediatrics, University of Pécs Medical School, Pécs, Hungary
| | - Anikó Ujfalusi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Szegedi
- Division of Pediatric Hematology-Oncology, Institute of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Lilla Györgyi Tiszlavicz
- Department of Paediatrics and Paediatric Health Care Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Katalin Bartyik
- Department of Paediatrics and Paediatric Health Care Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Krisztina Csanádi
- Hemato-Oncology Unit, Heim Pál Children's Hospital, Budapest, Hungary
| | - György Péter
- Hemato-Oncology Unit, Heim Pál Children's Hospital, Budapest, Hungary
| | - Réka Simon
- Hemato-Oncology and Stem Cell Transplantation Unit, Velkey László Children's Health Center, Miskolc, Hungary
| | - Péter Hauser
- Hemato-Oncology and Stem Cell Transplantation Unit, Velkey László Children's Health Center, Miskolc, Hungary
| | - Ágnes Kelemen
- Hemato-Oncology and Stem Cell Transplantation Unit, Velkey László Children's Health Center, Miskolc, Hungary
| | - Endre Sebestyén
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Jakab
- Hungarian Childhood Cancer Registry, Hungarian Pediatric Oncology Network, Budapest, Hungary
| | - András Matolcsy
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
- Department of Laboratory Medicine, Karolinska Institute, Solna, Sweden
| | - Csongor Kiss
- Division of Pediatric Hematology-Oncology, Institute of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Kovács
- Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | | | - Csaba Bödör
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Donát Alpár
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
11
|
Srinivasan S, Ramanathan S, Kumar S, Peyam S, Radhakrishnan V. Prevalence and prognostic significance of IKZF1 deletion in paediatric acute lymphoblastic leukemia: A systematic review and meta-analysis. Ann Hematol 2023:10.1007/s00277-023-05250-1. [PMID: 37154889 DOI: 10.1007/s00277-023-05250-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023]
Abstract
IKZF1 (IKAROS family Zinc Finger 1) alteration is an essential regulator of both T- and B-cell lineage specification with leukemogenic potential. IKZF1 deletion have been described in childhood acute lymphoblastic leukemia (ALL) with varying prevalence often influenced by underlying cytogenetics and also shown to have diverse prognostic significance. We aimed to evaluate the prevalence and prognostic significance of IKZF1 deletion among childhood ALL. Electronic databases of MEDLINE, EMBASE and SCOPUS were searched and 32 studies found eligible. Estimated prevalence of IKZF1 deletion among BCR::ABL1 negative and BCR::ABL1 positive ALL patients was 14% (95%CI:13-16%, I2 = 79%; 26 studies) and 63% (95%CI:59-68% I2 = 42%; 10 studies) respectively. Most common site of IKZF1 deletion was whole chromosome (exon1-8) deletion in 32.3% (95%CI: 23.8-40.7%) followed by exon 4-7 deletion in 28.6% (95%CI: 19.7-37.5%). A positive minimal residual disease at the end of induction was more common among patients with IKZF1 deletion, odds ratio: 3.09 (95%CI:2.3-4.16, I2 = 54%; 15 studies). Event-free survival and overall survival were significantly worse for IKZF1 deletion, hazard ratio (HR): 2.10 (95%CI:1.90-2.32, I2 = 28%; 31 studies) and HR: 2.38 (95%CI:1.93-2.93, I2 = 40; 15 studies) respectively. In summary, the current meta-analysis highlights the frequency of IKZF1 deletion and its negative impact on survival in childhood ALL. Further studies exploring the influence of IKZF1 deletion in the presence of classical cytogenetic and other copy number alterations would further help in characterising its prognostic role.
Collapse
Affiliation(s)
- Shyam Srinivasan
- Department of Pediatric Oncology, ACTREC/Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Parel, Mumbai, 400 012, Maharashtra, India.
| | - Subramaniam Ramanathan
- Department of Pediatric Oncology and BMT, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, United Kingdom
| | - Shathish Kumar
- Department of Anaesthesiology, Manipal Hospital Whitefield, Bangalore, India
| | - Srinivasan Peyam
- Department of Pediatrics, Pediatric Hematology-oncology Division, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | |
Collapse
|
12
|
Öfverholm I, Rezayee F, Heyman M, Harila A, Arvidsson L, Schmiegelow K, Norén-Nyström U, Barbany G. The prognostic impact of IKZF1 deletions and UKALL genetic classifiers in paediatric B-cell precursor acute lymphoblastic leukaemia treated according to NOPHO 2008 protocols. Br J Haematol 2023. [PMID: 37156607 DOI: 10.1111/bjh.18852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
We investigated 390 paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) patients treated according to NOPHO ALL 2008, regarding copy number alterations (CNA) of eight loci associated with adverse prognosis, including IKZF1. The impact on outcome was investigated for each locus individually, combined as CNA profiles and together with cytogenetic information. The presence of IKZF1 deletion or a poor-risk CNA profile was associated with poor outcome in the whole cohort. In the standard-risk group, IKZF1-deleted cases had an inferior probability of relapse-free survival (pRFS) (p ≤ 0.001) and overall survival (pOS) (p ≤ 0.001). Additionally, among B-other patients, IKZF1 deletion correlated with poor pRFS (60% vs. 90%) and pOS (65% vs. 89%). Both IKZF1 deletion and a poor-risk CNA profile were independent factors for relapse and death in multivariable analyses adjusting for known risk factors including measurable residual disease. Our data indicate that BCP-ALL patients with high-risk CNA or IKZF1 deletion have worse prognosis despite otherwise low-risk features. Conversely, patients with both a good CNA and cytogenetic profile had a superior relapse-free (p ≤ 0.001) and overall survival (p ≤ 0.001) in the cohort, across all risk groups. Taken together, our findings highlight the potential of CNA assessment to refine stratification in ALL.
Collapse
Affiliation(s)
- Ingegerd Öfverholm
- Department of Molecular Medicine and Surgery and Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Fatemah Rezayee
- Department of Molecular Medicine and Surgery and Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Mats Heyman
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Arja Harila
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Linda Arvidsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kjeld Schmiegelow
- Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
- Faculty of Medicine, Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Gisela Barbany
- Department of Molecular Medicine and Surgery and Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
13
|
Østergaard A, Enshaei A, Pieters R, Vora A, Horstmann MA, Escherich G, Johansson B, Heyman M, Schmiegelow K, Hoogerbrugge PM, den Boer ML, Kuiper RP, Moorman AV, Boer JM, van Leeuwen FN. The Prognostic Effect of IKZF1 Deletions in ETV6:: RUNX1 and High Hyperdiploid Childhood Acute Lymphoblastic Leukemia. Hemasphere 2023; 7:e875. [PMID: 37153875 PMCID: PMC10162793 DOI: 10.1097/hs9.0000000000000875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 03/09/2023] [Indexed: 05/10/2023] Open
Abstract
IKZF1 deletions are an established prognostic factor in childhood acute lymphoblastic leukemia (ALL). However, their relevance in patients with good risk genetics, namely ETV6::RUNX1 and high hyperdiploid (HeH), ALL remains unclear. We assessed the prognostic impact of IKZF1 deletions in 939 ETV6::RUNX1 and 968 HeH ALL patients by evaluating data from 16 trials from 9 study groups. Only 3% of ETV6::RUNX1 cases (n = 26) were IKZF1-deleted; this adversely affected survival combining all trials (5-year event-free survival [EFS], 79% versus 92%; P = 0.02). No relapses occurred among the 14 patients with an IKZF1 deletion treated on a minimal residual disease (MRD)-guided protocols. Nine percent of HeH cases (n = 85) had an IKZF1 deletion; this adversely affected survival in all trials (5-year EFS, 76% versus 89%; P = 0.006) and in MRD-guided protocols (73% versus 88%; P = 0.004). HeH cases with an IKZF1 deletion had significantly higher end of induction MRD values (P = 0.03). Multivariate Cox regression showed that IKZF1 deletions negatively affected survival independent of sex, age, and white blood cell count at diagnosis in HeH ALL (hazard ratio of relapse rate [95% confidence interval]: 2.48 [1.32-4.66]). There was no evidence to suggest that IKZF1 deletions affected outcome in the small number of ETV6::RUNX1 cases in MRD-guided protocols but that they are related to higher MRD values, higher relapse, and lower survival rates in HeH ALL. Future trials are needed to study whether stratifying by MRD is adequate for HeH patients or additional risk stratification is necessary.
Collapse
Affiliation(s)
- Anna Østergaard
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Amir Enshaei
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Ajay Vora
- Department of Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Martin A. Horstmann
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- University Medical Center Hamburg, Research Institute Children’s Cancer Center, Hamburg, Germany
| | - Gabriele Escherich
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bertil Johansson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Sweden
- Department of Clinical Genetics, Pathology, and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Mats Heyman
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Denmark
| | | | - Monique L. den Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Roland P. Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Department of Genetics, University Medical Center Utrecht, Netherlands
| | - Anthony V. Moorman
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Judith M. Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | | |
Collapse
|
14
|
Eadie LN, Rehn JA, Breen J, Osborn MP, Jessop S, Downes CEJ, Heatley SL, McClure BJ, Yeung DT, Revesz T, Saxon B, White DL. Case Report: Rare IKZF1 Gene Fusions Identified in Neonate with Congenital KMT2A-Rearranged Acute Lymphoblastic Leukemia. Genes (Basel) 2023; 14:genes14020264. [PMID: 36833191 PMCID: PMC9956107 DOI: 10.3390/genes14020264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Chromosomal rearrangements involving the KMT2A gene occur frequently in acute lymphoblastic leukaemia (ALL). KMT2A-rearranged ALL (KMT2Ar ALL) has poor long-term survival rates and is the most common ALL subtype in infants less than 1 year of age. KMT2Ar ALL frequently occurs with additional chromosomal abnormalities including disruption of the IKZF1 gene, usually by exon deletion. Typically, KMT2Ar ALL in infants is accompanied by a limited number of cooperative le-sions. Here we report a case of aggressive infant KMT2Ar ALL harbouring additional rare IKZF1 gene fusions. Comprehensive genomic and transcriptomic analyses were performed on sequential samples. This report highlights the genomic complexity of this particular disease and describes the novel gene fusions IKZF1::TUT1 and KDM2A::IKZF1.
Collapse
Affiliation(s)
- Laura N. Eadie
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Jacqueline A. Rehn
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - James Breen
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- South Australian Genomics Centre (SAGC), Adelaide, SA 5000, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Michael P. Osborn
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australian & New Zealand Children’s Haematology/Oncology Group, Clayton, VIC 3168, Australia
- Australasian Leukaemia & Lymphoma Group, Richmond, VIC 3121, Australia
- Department of Haematology & Oncology, Women’s & Children’s Hospital, Adelaide, SA 5000, Australia
- Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Sophie Jessop
- Australian & New Zealand Children’s Haematology/Oncology Group, Clayton, VIC 3168, Australia
- Department of Haematology & Oncology, Women’s & Children’s Hospital, Adelaide, SA 5000, Australia
| | - Charlotte E. J. Downes
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Sciences, Engineering and Technology, University of Adelaide, Adelaide, SA 5000, Australia
| | - Susan L. Heatley
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australian & New Zealand Children’s Haematology/Oncology Group, Clayton, VIC 3168, Australia
| | - Barbara J. McClure
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - David T. Yeung
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia & Lymphoma Group, Richmond, VIC 3121, Australia
- Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Tamas Revesz
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australian & New Zealand Children’s Haematology/Oncology Group, Clayton, VIC 3168, Australia
- Department of Haematology & Oncology, Women’s & Children’s Hospital, Adelaide, SA 5000, Australia
| | - Benjamin Saxon
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Department of Haematology & Oncology, Women’s & Children’s Hospital, Adelaide, SA 5000, Australia
- Clinical Services and Research Division, Australian Red Cross Blood Service, Adelaide, SA 5000, Australia
| | - Deborah L. White
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australian & New Zealand Children’s Haematology/Oncology Group, Clayton, VIC 3168, Australia
- Australasian Leukaemia & Lymphoma Group, Richmond, VIC 3121, Australia
- Faculty of Sciences, Engineering and Technology, University of Adelaide, Adelaide, SA 5000, Australia
- Australian Genomics Health Alliance, Parkville, VIC 3052, Australia
- Correspondence:
| |
Collapse
|
15
|
Felice MS, Rubio PL, Digiorge J, Barreda Frank M, Martínez CS, Guitter MR, Sajaroff EO, Sánchez La Rosa CG, Pennella CL, Peruzzo LB, Deu MA, Alfaro EM, Guardia MC, Gutierrez G, Fernández Barbieri MA, Recondo E, Vides Herrera MS, Livio V, Arnaiz C, Romero C, Alonso CN, Rossi JG. Impact of IKZF1 Deletions in the Prognosis of Childhood Acute Lymphoblastic Leukemia in Argentina. Cancers (Basel) 2022; 14:cancers14133283. [PMID: 35805054 PMCID: PMC9266042 DOI: 10.3390/cancers14133283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 12/23/2022] Open
Abstract
An association of deletions in the IKZF1 gene (IKZF1del) with poor prognosis in acute lymphoblastic leukemia (ALL) has been demonstrated. Additional deletions in other genes (IKZF1plus) define different IKZF1del subsets. We analyzed the influence of IKZF1del and/or IKZF1plus in the survival of children with ALL. From October 2009 to July 2021, 1055 bone marrow samples from patients with ALL were processed by Multiplex ligation-dependent probe amplification (MLPA). Of them, 28 patients died during induction and 4 were lost-in-follow-up, resulting in an eligible 1023 cases. All patients were treated according to ALLIC-BFM-2009-protocol. Patients were classified into three subsets: IKZF1not-deleted (IKZFF1not-del), IKZF1deleted (IKZF1del) and IKZF1del plus deletion of PAX5, CDKN2A, CDKN2B and/or alterations in CRLF2 with ERG-not-deleted (IKZF1plus). The LFSp and SE were calculated with the Kaplan−Meier calculation and compared with a log-rank test. From the 1023 eligible patients, 835 (81.6%) were defined as IKZF1not-del, 94 (9.2%) as IKZF1del and 94 (9.2%) as IKZF1plus. Of them, 100 (9.8%) corresponded to Standard-Risk (SRG), 629 (61.5%) to Intermediate-Risk (IRG) and 294 (28.7%) to High-Risk (HRG) groups. LFSp(SE) was 7 5(2)% for IKZF1not-del, 51 (6)% for IKZF1del and 48 (6)% for IKZF1plus (p-value < 0.00001). LFSp(SE) according to the risk groups was: in SRG, 91 (4)% for IKZF1not-del, 50 (35)% IKZF1del and 100% IKZF1plus (p-value = ns); in IRG, 77 (2)% IKZF1not-del, 61 (10)% IKZF1del and 54 (7)% IKZF1plus (p-value = 0.0005) and in HRG, 61 (4)% IKZF1not-del, 38 (8)% IKZF1del and 35 (9)% IKZF1plus (p-value = 0.0102). The IKZF1 status defines a population of patients with a poor outcome, mainly in IRG. No differences were observed between IKZF1del versus IKZF1plus. MLPA studies should be incorporated into the risk-group stratification of pediatric ALL.
Collapse
Affiliation(s)
- María Sara Felice
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
- Correspondence:
| | - Patricia Laura Rubio
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - Jorge Digiorge
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - Mariángeles Barreda Frank
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - Celeste Sabrina Martínez
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - Myriam Ruth Guitter
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - Elisa Olga Sajaroff
- Immunology and Rheumatology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (E.O.S.); (J.G.R.)
| | - Cristian Germán Sánchez La Rosa
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - Carla Luciana Pennella
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - Luisina Belén Peruzzo
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - María Alejandra Deu
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - Elizabeth Melania Alfaro
- Hematology and Oncology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (P.L.R.); (J.D.); (M.B.F.); (C.S.M.); (M.R.G.); (C.G.S.L.R.); (C.L.P.); (L.B.P.); (M.A.D.); (E.M.A.)
| | - María Constanza Guardia
- Hematology and Oncology Department, Hospital del Niño Jesús, San Miguel de Tucumán, Tucumán 4000, Argentina;
| | - Gladys Gutierrez
- Hematology and Oncology Department, Hospital Juan Pablo II, Corrientes 1435, Argentina;
| | | | - Ezequiel Recondo
- Hematology and Oncology Department, Hospital Nacional de Clínicas San Martín, Buenos Aires 1245, Argentina;
| | | | - Vanina Livio
- Hematology and Oncology Department, Hospital Avelino Castelán, Resistencia, Chaco 3508, Argentina;
| | - Constanza Arnaiz
- Hematology and Oncology Department, Hospital de Niños Castro Rendón, Neuquén 8300, Argentina;
| | - Carolina Romero
- Hematology and Oncology Department, Hospital Alexander Fleming OSEP, Mendoza 5500, Argentina;
| | - Cristina Noemi Alonso
- Area of Specialized Laboratories, Hospital de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires 1245, Argentina;
| | - Jorge Gabriel Rossi
- Immunology and Rheumatology Department, Hospital de Pediatría Juan P. Garrahan, Buenos Aires 1245, Argentina; (E.O.S.); (J.G.R.)
| |
Collapse
|
16
|
Zhang J, Xu XJ, Liu L, Song H, Shen H, Xu W, Zhao F, Liang J, Liao C, Wang Y, Xia T, Cao S, Tang Y, Qin J, Shen D. Clinical and Genetic Characteristics of IKZF1 Mutation in Chinese Children With B-Cell Acute Lymphoblastic Leukemia. Front Genet 2022; 13:822832. [PMID: 35419036 PMCID: PMC9000999 DOI: 10.3389/fgene.2022.822832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/22/2022] [Indexed: 11/21/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a malignancy associated with altered lymphoid precursor hyperplasia and accompanied with different genetic mutations. Few studies have been reported on the association between gene mutations and clinical features of IKZF1 mutation in children with B-cell ALL (B-ALL). We investigated clinical and genetic characteristics in 200 newly diagnosed pediatric B-ALL through multiplex ligation-dependent probe amplification (MLPA) and targeted next-generation sequencing (NGS) method. We found that IKZF1 mutations, including large segment deletions, small insertions or deletions (InDels) and single nucleotide variations (SNVs), were detected in 22 patients with a positive mutation rate of 11.0%. IKZF1 mutation was significantly associated with higher WBC count (19.38 × 109/L vs. 5.80 × 109/L, p = 0.002). Compared with IKZF1 wild-type cases, a higher frequency of IL7R gene mutation was discovered in IKZF1 mutant cases (9.1% vs. 0.0%, p = 0.012). Patients with IKZF1 mutation were less sensitive to glucocorticoid induction than patients without IKZF1 mutation (63.6% vs. 9.0%, p < 0.001). On the 15th day of induction, minimal residual disease (MRD) > 10−3 level were higher in IKZF1 mutant patients than wild-type patients (45.5% vs. 22.3%, p = 0.018). In conclusion, our study reveals the association between genetic mutations and clinical features in Chinese children with B-ALL, which might contribute to molecular classification, risk stratification and prognosis evaluation, and provide new ideas for targeted therapy in ALL.
Collapse
Affiliation(s)
- Jingying Zhang
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiao-Jun Xu
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Lixia Liu
- Acornmed Biotechnology Co., Ltd., Tianjin, China
| | - Hua Song
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Heping Shen
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Weiqun Xu
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Fenying Zhao
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Juan Liang
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Chan Liao
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Yan Wang
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Tian Xia
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Shanbo Cao
- Acornmed Biotechnology Co., Ltd., Tianjin, China
| | - Yongmin Tang
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiayue Qin
- Acornmed Biotechnology Co., Ltd., Tianjin, China
| | - Diying Shen
- Division/Center of Pediatric Hematology-Oncology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.,The Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
17
|
Braun M, Pastorczak A, Sedek L, Taha J, Madzio J, Jatczak-Pawlik I, Wypyszczak K, Matysiak M, Derwich K, Lejman M, Kazanowska B, Szczepanski T, Kowalczyk JR, Mlynarski W. Prognostic significance of IKZF1 deletions and IKZF1 plus profile in children with B-cell precursor acute lymphoblastic leukemia treated according to the ALL-IC BFM 2009 protocol. Hematol Oncol 2022; 40:430-441. [PMID: 35118711 DOI: 10.1002/hon.2973] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/07/2022]
Abstract
The strongest predictors of outcome in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL) are minimal residual disease (MRD) and specific molecular abnormalities. One unfavorable prognostic factor is the presence of IKZF1 gene aberrations, particularly when co-occurring with high MRD level at the end of induction treatment. The present study determines the predictive value of a recently-defined IKZF1-plus (IKZF1plus ) microdeletion profile in 373 children with BCP-ALL treated according to the ALL-IC BFM 2009 protocol. IKZF1-wild type (IKZF1wt ) patients demonstrated lower leukemic burden parameters than those carrying IKZF1 deletion (IKZF1del [n=26, 7.0%]) or IKZF1plus pattern (n=34, 9.1%): i) median blast percentage at diagnosis (78.0% vs. 86.9% vs. 86.0%; p=0.021); ii) median MRD level at day 15 of induction protocol (0.3% vs. 2.1% vs. 0.8%; p=0.011); iii) poor steroid response (7.6% vs. 26.5% vs. 12.5%; p=0.010). MRD level at day 33 (MRD33) exceeding 10-4 was more frequently observed in both the IKZF1del and IKZF1plus subgroups than in IKZF1wt patients (n=9 [36.0%] vs. n=13 [41.9%] vs. n=70 [24.0%], p=0.051). IKZF1plus individuals showed a tendency for a lower MRD reduction between day 15 and 33 compared to IKZF1del patients (p=0.124). IKZF1del and IKZF1plus patients showed decreased relapse-free survival (HR [95%CI] for IKZF1wt as reference=2.72 [1.21-6.11] and 2.00 [0.87-4.49], respectively, p=0.023). Both genetic markers including IKZF1del and IKZF1plus microdeletion profile provide additional predictive value of treatment outcome in childhood BCP-ALL and may contribute to more efficient patient stratification; the same is true in MRD guided protocols, which are based on flow cytometric measurements on day 15 of induction protocol. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Marcin Braun
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Poland.,Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Poland
| | - Agata Pastorczak
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Poland
| | - Lukasz Sedek
- Department of Pediatric Hematology and Oncology, Medical University of Silesia, Zabrze, Poland
| | - Joanna Taha
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Poland
| | - Joanna Madzio
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Poland
| | | | - Kamila Wypyszczak
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Poland
| | - Michał Matysiak
- Department of Pediatric Hematology and Oncology, Medical University of Warsaw, Poland
| | - Katarzyna Derwich
- Department of Pediatric Hematology, Oncology and Transplantology, University of Medical Sciences, Poznan, Poland
| | - Monika Lejman
- Laboratory of Genetic Diagnostics, Medical University of Lublin, Poland
| | - Bernarda Kazanowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Wroclaw, Poland
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Medical University of Silesia, Zabrze, Poland
| | - Jerzy R Kowalczyk
- Department of Pediatric Hematology and Oncology, Medical University of Lublin, Poland
| | - Wojciech Mlynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Poland
| |
Collapse
|
18
|
Wu X, Liu M, Wang Q. IKZF1 Rs4132601 Polymorphism and Susceptibility to Acute Lymphocytic Leukemia in Children: A Meta-analysis. J Pediatr Hematol Oncol 2022; 44:e101-e108. [PMID: 33661176 DOI: 10.1097/mph.0000000000002130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/27/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Many studies have shown that IKAROS family zinc finger 1 (IKZF1) rs4132601 polymorphism is strongly linked to acute lymphoblastic leukemia (ALL) in children, but their conclusions have been inconsistent. OBJECTIVE This meta-analysis is set out to investigate the association between IKZF1 rs4132601 polymorphism and its susceptibility to childhood ALL. DATA AND METHODS On the basis of inclusion criteria, PubMed, EMBASE, Web of Science, CNKI, China Wanfang, VIP, and other databases were searched from the time of the establishment of the library database to December 2019 for all case-control studies. Stata 15.0 was applied for meta-analysis to calculate the combined odds ratio (OR) value and 95% confidence interval (CI) of each genotype at IKZF1 rs4132601. Subgroup analysis done by ethnicity, sensitivity analysis, and publication bias assessment was further performed. RESULTS Nine pieces of literature was included in this meta-analysis, including 2281 children with ALL and 2923 controls. There were significant differences in the allelic model (T vs. G: combined OR=0.75, 95% CI: 0.68-0.82, P<0.05) in both Asian and Caucasian children. In addition to this, there were statistically significant differences in the dominant, homozygous and heterozygous genetic model in both Asian and Caucasian children. The difference was significant in the recessive genetic model (TT vs. TG+GG: combined OR=0.75, 95% CI: 0.67-0.84) in Caucasian children, but not in Asian children (combined OR=0.85, 95% CI: 0.70-1.04, P>0.05). CONCLUSION There is a strong correlation between IKZF1 rs4132601 polymorphism and ALL in children. Compared with the G allele, T alleles can lower the risk of childhood ALL, and TT, TT+TG and TG genotypes can also reduce the risk of ALL in children.
Collapse
Affiliation(s)
- Xue Wu
- Departments of Pediatric Preventive Health
| | - Mengyi Liu
- Departments of Pediatric Preventive Health
| | - Qin Wang
- Pediatrics, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| |
Collapse
|
19
|
Lee SHR, Li Z, Tai ST, Oh BLZ, Yeoh AEJ. Genetic Alterations in Childhood Acute Lymphoblastic Leukemia: Interactions with Clinical Features and Treatment Response. Cancers (Basel) 2021; 13:4068. [PMID: 34439222 PMCID: PMC8393341 DOI: 10.3390/cancers13164068] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/03/2021] [Accepted: 08/08/2021] [Indexed: 12/28/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer among children. This aggressive cancer comprises multiple molecular subtypes, each harboring a distinct constellation of somatic, and to a lesser extent, inherited genetic alterations. With recent advances in genomic analyses such as next-generation sequencing techniques, we can now clearly identify >20 different genetic subtypes in ALL. Clinically, identifying these genetic subtypes will better refine risk stratification and determine the optimal intensity of therapy for each patient. Underpinning each genetic subtype are unique clinical and therapeutic characteristics, such as age and presenting white blood cell (WBC) count. More importantly, within each genetic subtype, there is much less variability in treatment response and survival outcomes compared with current risk factors such as National Cancer Institute (NCI) criteria. We review how this new taxonomy of genetic subtypes in childhood ALL interacts with clinical risk factors used widely, i.e., age, presenting WBC, IKZF1del, treatment response, and outcomes.
Collapse
Affiliation(s)
- Shawn H. R. Lee
- VIVA-University Children’s Cancer Centre, Khoo-Teck Puat-National University Children’s Medical Institute, National University Hospital, Singapore 119074, Singapore; (S.H.R.L.); (B.L.Z.O.)
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore; (Z.L.); (S.T.T.)
| | - Zhenhua Li
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore; (Z.L.); (S.T.T.)
| | - Si Ting Tai
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore; (Z.L.); (S.T.T.)
| | - Bernice L. Z. Oh
- VIVA-University Children’s Cancer Centre, Khoo-Teck Puat-National University Children’s Medical Institute, National University Hospital, Singapore 119074, Singapore; (S.H.R.L.); (B.L.Z.O.)
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore; (Z.L.); (S.T.T.)
| | - Allen E. J. Yeoh
- VIVA-University Children’s Cancer Centre, Khoo-Teck Puat-National University Children’s Medical Institute, National University Hospital, Singapore 119074, Singapore; (S.H.R.L.); (B.L.Z.O.)
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore; (Z.L.); (S.T.T.)
| |
Collapse
|
20
|
Gupta SK, Bakhshi S, Kamal VK, Gupta R, Sharma P, Pushpam D, Sahoo RK, Sharma A. Proposal and clinical application of molecular genetic risk scoring system, "MRplus", for BCR-ABL1 negative pediatric B-cell acute lymphoblastic leukemia- report from a single centre. Leuk Res 2021; 111:106683. [PMID: 34371436 DOI: 10.1016/j.leukres.2021.106683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/28/2021] [Accepted: 08/01/2021] [Indexed: 12/25/2022]
Abstract
INTRODUCTION We propose "MRplus", a molecular genetic risk score and check its clinical application in the risk-stratification of pediatric B-ALL. METHODS The genomic DNA of untreated pediatricBCR-ABL1 negative B-ALL patients was analyzed for deletions of IKZF1, PAX5, CDKN2A/B, BTG1, RB1, ETV6, EBF1, ERG, pseudoautosomal region(PAR) genes using multiplex ligation-dependent probe amplification, along with the routine genetic work-up. The patients were assigned an 'M'score- 0 (M0) for low and 1 (M1) for high genetic-risk as per the criteria by Moorman et al., and another score "IKplus"-1 (IKplus1) for IKZF1plus as per the criteria by Stanulla et al., and 0 (IKplus0) for other patients. The final "MRplus" risk-score of 0 (MRplus0), 1 (MRplus1) or 2 (MRplus2) was obtained by adding both these scores. The association of risk scores with overall survival (OS) and event free survival(EFS) was seen using Cox proportion hazard model. The overall goodness of fit of the model was done using Cox-Snell residuals. RESULTS The median age of 320 patients was 6 years (1-18 years). The patients with score M1 were 139 (43.4 %), M0-181 (56.6 %); IKplus1-32 (10 %) and IKplus0-288 (90 %). The final "MRplus" score of 0,1,or 2 was obtained in 181(56.6 %), 107(33.4 %) and 32(10 %) patients respectively. The post-induction remission rate was 90.7 %, 77.8 %, 73.9 % (p = 0.004); 4-year OS 67 %, 48 %, 27 % (p < 0.001); and 4-year EFS 56 %, 34 %, 19 %(p < 0.001) in patients with "MRplus" score 0,1,and 2 respectively. CONCLUSIONS The proposed "MRplus" scoring at baseline could identify three distinct risk groups-good (MRplus0), intermediate (MRplus1) and poor (MRplus2), with different outcomes; in pediatricBCR-ABL1 negative B-ALL. This may help in better risk-stratification and selection of patients for alternative treatment approaches.
Collapse
Affiliation(s)
- Sanjeev Kumar Gupta
- Laboratory Oncology Unit, Dr BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Vineet Kumar Kamal
- Division of Epidemiology & Biostatistics, ICMR National Institute of Epidemiology, Chennai, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Preity Sharma
- Laboratory Oncology Unit, Dr BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Deepam Pushpam
- Department of Medical Oncology, Dr BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Atul Sharma
- Department of Medical Oncology, Dr BRA IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
21
|
Huang YJ, Liu HC, Jaing TH, Wu KH, Wang SC, Yen HJ, Hsiao CC, Chen SH, Lin PC, Yeh TC, Sheen JM, Chen YC, Chang TK, Huang FL, Chao YH, Hou JY, Yang CP, Lin TH, Shih LY. RAS pathway mutation is an added-value biomarker in pediatric Philadelphia-negative B-cell acute lymphoblastic leukemia with IKZF1 deletions. Pediatr Blood Cancer 2021; 68:e28899. [PMID: 33522704 DOI: 10.1002/pbc.28899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/26/2020] [Indexed: 11/12/2022]
Abstract
BACKGROUND IKZF1deletion is an unfavorable factor in Philadelphia negative (Ph -) B-cell acute lymphoblastic leukemia. However, the effects of IKZF1 deletions co-existing genetic alterations in Ph (-) ALL have not been extensively studied. METHODS Bone marrow samples from 368 children with Ph (-) ALL were analyzed by using multiplex ligation-dependent probe amplification kit for detection of gene deletions and Sanger sequencing for mutational analysis of RAS pathway genes. The outcome was analyzed on 215 patients treated with Taiwan Pediatric Oncology Group-ALL-2002 protocol. RESULTS IKZF1 deletions were present in 12.8% and IKZF1plus in 6.3% of patients. Mutations of RAS pathway genes were detected in 25.0% of IKZF1-deleted patients. The 10-year event-free survival (EFS) of IKZF1-undeleted patients was significantly better compared with IKZF1-deleted patients (80.0% vs. 47.8%, p = 0.001). Compared with outcome of patients harboring IKZF1 deletion alone, no difference in EFS was observed in patients with IKZF1plus , whereas three patients carried both IKZF1 and ERG deletions had a superior 10-year EFS (100%). The 10-year EFS of patients with any gene mutation of RAS pathway was worse than that of patients with wild-type genes (79.1% vs. 61.6%, p = 0.033). In multivariate analysis, RAS pathway mutations and IKZF1 deletion were independent predictors of inferior EFS. Co-existence of IKZF1 deletion with RAS pathway mutations had a worst 10-year EFS (11.1 ± 10.5%) and 10-year OS (53.3 ± 17.6%). CONCLUSIONS Our results showed that RAS pathway mutation is an added-value biomarker in pediatric IKZF1-deleted Ph (-) ALL patients.
Collapse
Affiliation(s)
- Ying-Jung Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hsi-Che Liu
- Department of Hematology-Oncology, Mackay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Tang-Her Jaing
- Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kang-Hsi Wu
- Division of Pediatric Hematology and Oncology, China Medical University Children's Hospital, Taichung, Taiwan
| | - Shih-Chung Wang
- Division of Pediatric Hematology-Oncology, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Hsiu-Ju Yen
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Cheng Hsiao
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Chang Gung Memorial Hospital at Kaohsiung, Kaohsiung, Taiwan
| | - Shih-Hsiang Chen
- Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pei-Chin Lin
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ting-Chi Yeh
- Department of Hematology-Oncology, Mackay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Jiunn-Ming Sheen
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Chang Gung Memorial Hospital at Kaohsiung, Kaohsiung, Taiwan.,Department of Pediatrics, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Yu-Chieh Chen
- Department of Pediatrics, Chang Gung Memorial Hospital at Kaohsiung, Kaohsiung, Taiwan
| | - Te-Kau Chang
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Fang-Liang Huang
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yu-Hua Chao
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jen-Yin Hou
- Department of Hematology-Oncology, Mackay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Chao-Ping Yang
- Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan
| | - Tung-Huei Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Lee-Yung Shih
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
22
|
Gupta SK, Bakhshi S, Gupta R, Sharma P, Pushpam D, Sahoo RK, Kamal VK. IKZF1 Deletion Subtyping and Outcome Analysis in BCR-ABL1-Negative Pediatric B-Cell Acute Lymphoblastic Leukemia: A Single-Institution Experience from North India. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:e666-e673. [PMID: 33906825 DOI: 10.1016/j.clml.2021.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/12/2021] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND IKZF1 deletions are associated with adverse outcomes in B-cell acute lymphoblastic leukemia (B-ALL). We assessed the prevalence and clinical impact of functional subtypes of IKZF1 deletions in pediatric BCR-ABL1-negative B-ALL. PATIENTS AND METHODS This retrospective study of IKZF1 deletions was done in cases of pediatric BCR-ABL1-negative B-ALL. The genomic DNA of cases, over a 53-month period, was analyzed using multiplex ligation-dependent probe amplification and multiplex fluorescent polymerase chain reaction. The deletions were divided into functional subgroups: (1) loss-of-function/haploinsufficiency, (2) dominant-negative, and (3) a combination of both types of deletion. The post-induction remission status, event-free survival (EFS), and overall survival (OS) were noted. RESULTS Out of 320 cases, 47 (14.7%) had IKZF1 deletions. Thirty-six of the 47 (77%) had loss-of-function deletions, 10 (21%) had dominant-negative deletions, and one (2%) had a combination of both types. The post-induction remission rates in cases with loss-of-function deletions (22/30, 73%; P = .060) and dominant-negative deletions (4/5, 80%; P = .517) were lower compared with those without deletions (215/248, 86.7%). These cases also had worse median EFS: 21.1 months (P = .006) for loss-of-function and 15.4 months (P = .156) for dominant-negative deletions, compared with 46.4 months in cases without IKZF1 deletions. They also had worse median OS: 23.4 months (P = .012) for loss-of-function deletions and 15.7 months (P = .233) for dominant-negative deletions, compared with median not reached in cases without IKZF1 deletions. CONCLUSION The IKZF1 deletions were seen in 14.7% of BCR-ABL1-negative pediatric B-ALL. Most of these deletions (77%) were loss-of-function type. The cases with loss-of-function deletions had lower remission rates and poor EFS and OS compared with cases without IKZF1 deletions. A similar trend of poor outcome was seen in the few cases with dominant-negative IKZF1 deletions.
Collapse
Affiliation(s)
- Sanjeev Kumar Gupta
- Laboratory Oncology Unit, Dr. B.R.A. IRCH, All India Institute of Medical Sciences, New Delhi, India.
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B.R.A. IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr. B.R.A. IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Preity Sharma
- Laboratory Oncology Unit, Dr. B.R.A. IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Deepam Pushpam
- Department of Medical Oncology, Dr. B.R.A. IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr. B.R.A. IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Vineet Kumar Kamal
- Division of Epidemiology and Biostatistics, National Institute of Epidemiology, Chennai, India
| |
Collapse
|
23
|
IKAROS and CK2 regulate expression of BCL-XL and chemosensitivity in high-risk B-cell acute lymphoblastic leukemia. Blood 2021; 136:1520-1534. [PMID: 32396934 DOI: 10.1182/blood.2019002655] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 04/23/2020] [Indexed: 12/18/2022] Open
Abstract
High-risk B-cell acute lymphoblastic leukemia (B-ALL) is an aggressive disease, often characterized by resistance to chemotherapy. A frequent feature of high-risk B-ALL is loss of function of the IKAROS (encoded by the IKZF1 gene) tumor suppressor. Here, we report that IKAROS regulates expression of the BCL2L1 gene (encodes the BCL-XL protein) in human B-ALL. Gain-of-function and loss-of-function experiments demonstrate that IKAROS binds to the BCL2L1 promoter, recruits histone deacetylase HDAC1, and represses BCL2L1 expression via chromatin remodeling. In leukemia, IKAROS' function is impaired by oncogenic casein kinase II (CK2), which is overexpressed in B-ALL. Phosphorylation by CK2 reduces IKAROS binding and recruitment of HDAC1 to the BCL2L1 promoter. This results in a loss of IKAROS-mediated repression of BCL2L1 and increased expression of BCL-XL. Increased expression of BCL-XL and/or CK2, as well as reduced IKAROS expression, are associated with resistance to doxorubicin treatment. Molecular and pharmacological inhibition of CK2 with a specific inhibitor CX-4945, increases binding of IKAROS to the BCL2L1 promoter and enhances IKAROS-mediated repression of BCL2L1 in B-ALL. Treatment with CX-4945 increases sensitivity to doxorubicin in B-ALL, and reverses resistance to doxorubicin in multidrug-resistant B-ALL. Combination treatment with CX-4945 and doxorubicin show synergistic therapeutic effects in vitro and in preclinical models of high-risk B-ALL. Results reveal a novel signaling network that regulates chemoresistance in leukemia. These data lay the groundwork for clinical testing of a rationally designed, targeted therapy that combines the CK2 inhibitor, CX-4945, with doxorubicin for the treatment of hematopoietic malignancies.
Collapse
|
24
|
Allogeneic HCT for adults with B-cell precursor acute lymphoblastic leukemia harboring IKZF1 gene mutations. A study by the Acute Leukemia Working Party of the EBMT. Bone Marrow Transplant 2020; 56:1047-1055. [PMID: 33235351 DOI: 10.1038/s41409-020-01139-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 10/03/2020] [Accepted: 11/06/2020] [Indexed: 11/08/2022]
Abstract
The presence of IKZF1 gene mutations is associated with poor prognosis of B-cell precursor acute lymphoblastic leukemia (BCP-ALL). The goal of this retrospective study was to evaluate outcome of allogeneic hematopoietic cell transplantation (allo-HCT) in this population. Ninety-five patients transplanted in first (n = 75) or second (n = 20) complete remission (CR) from either HLA-matched sibling (n = 32), unrelated (n = 47) or haploidentical (n = 16) donor were included in the analysis. The probabilities of the overall survival (OS) and leukemia-free survival (LFS) at 2 years were 55% and 47%, respectively. Relapse incidence (RI) was 32% while non-relapse mortality (NRM), 21%. The incidence of grade II-IV acute graft-versus-host disease (GVHD) and chronic GVHD was 34% and 30%, respectively. The probability of GVHD and relapse-free survival (GRFS) was 35%. In a multivariate analysis positive minimal residual disease (MRD) status was associated with decreased chance of LFS (HR = 3.15, p = 0.004) and OS (HR = 2.37, p = 0.049) as well as increased risk of relapse (HR = 5.87, p = 0.003). Disease stage (CR2 vs. CR1) affected all, LFS, OS, GRFS, RI, and NRM. Results of allo-HCT for patients with BCP-ALL and IKZF1 mutations are generally improving, however, individuals with detectable MRD have poor prognosis and require additional intervention prior to transplantation.
Collapse
|
25
|
MLPA and DNA index improve the molecular diagnosis of childhood B-cell acute lymphoblastic leukemia. Sci Rep 2020; 10:11501. [PMID: 32661308 PMCID: PMC7359332 DOI: 10.1038/s41598-020-68311-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
Aneuploidy occurs within a significant proportion of childhood B-cell acute lymphoblastic leukemia (B-ALL). Some copy number variations (CNV), associated with novel subtypes of childhood B-ALL, have prognostic significance. A total of 233 childhood B-ALL patients were enrolled into this study. Focal copy number alterations of ERG, IKZF1, PAX5, ETV6, RB1, BTG1, EBF1, CDKN2A/2B, and the Xp22.33/Yp11.31 region were assessed by Multiplex Ligation-dependent Probe Amplification (MLPA). The MLPA telomere kit was used to identify aneuploidy through detection of whole chromosome loss or gain. We carried out these procedures alongside measurement of DNA index in order to identify, aneuploidy status in our cohort. MLPA telomere data and DNA index correlated well with aneuploidy status at higher sensitivity than cytogenetic analysis. Three masked hypodiploid patients, undetected by cytogenetics, and their associated copy number neutral loss of heterozygosity (CN-LOH) were identified by STR and SNP arrays. Rearrangements of TCF3, located to 19p, were frequently associated with 19p deletions. Other genetic alterations including iAMP21, IKZF1 deletions, ERG deletions, PAX5AMP, which have clinical significance or are associated with novel subtypes of ALL, were identified. In conclusion, appropriate application of MLPA aids the identifications of CNV and aneuploidy in childhood B-ALL.
Collapse
|
26
|
IKZF1 deletions in pediatric acute lymphoblastic leukemia: still a poor prognostic marker? Blood 2020; 135:252-260. [PMID: 31821407 DOI: 10.1182/blood.2019000813] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 11/21/2019] [Indexed: 12/31/2022] Open
Abstract
Improved personalized adjustment of primary therapy to the perceived risk of relapse by using new prognostic markers for treatment stratification may be beneficial to patients with acute lymphoblastic leukemia (ALL). Here, we review the advances that have shed light on the role of IKZF1 aberration as prognostic factor in pediatric ALL and summarize emerging concepts in this field. Continued research on the interplay of disease biology with exposure and response to treatment will be key to further improve treatment strategies.
Collapse
|
27
|
Vairy S, Tran TH. IKZF1 alterations in acute lymphoblastic leukemia: The good, the bad and the ugly. Blood Rev 2020; 44:100677. [PMID: 32245541 DOI: 10.1016/j.blre.2020.100677] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Advances in genomics have deepened our understanding of the biology of acute lymphoblastic leukemia (ALL), defined novel molecular leukemia subtypes, discovered new prognostic biomarkers and paved the way to emerging molecularly targeted therapeutic avenues. Since its discovery, IKZF1 has generated significant interest within the leukemia scientific community.IKZF1 plays a critical role in lymphoid development and its alterations cooperate to mediate leukemogenesis. IKZF1 alterations are present in approximately 15% of childhood ALL, rise in prevalence among adults with ALL and become highly enriched within kinase-driven ALL. A cumulating body of literature has highlighted the adverse prognostic impact of IKZF1 alterations in both Philadelphia chromosome (Ph)-negative and Ph-driven ALL. IKZF1 alterations thus emerge as an important prognostic biomarker in ALL. This article aims to provide a state-of-the-art review focusing on the prognostic clinical relevance of IKZF1 alterations in ALL, as well as current and future therapeutic strategies targeting IKZF1-altered ALL.
Collapse
Affiliation(s)
- Stephanie Vairy
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, Montréal, Québec, Canada
| | - Thai Hoa Tran
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, Montréal, Québec, Canada.
| |
Collapse
|
28
|
Abstract
Transcription factor IKZF1 (IKAROS) acts as a critical regulator of lymphoid differentiation and is frequently deleted or mutated in B-cell precursor acute lymphoblastic leukemia. IKZF1 gene defects are associated with inferior treatment outcome in both childhood and adult B-cell precursor acute lymphoblastic leukemia and occur in more than 70% of BCR-ABL1-positive and BCR-ABL1-like cases of acute lymphoblastic leukemia. Over the past few years, much has been learned about the tumor suppressive function of IKZF1 during leukemia development and the molecular pathways that relate to its impact on treatment outcome. In this review, we provide a concise overview on the role of IKZF1 during normal lymphopoiesis and the pathways that contribute to leukemia pathogenesis as a consequence of altered IKZF1 function. Furthermore, we discuss different mechanisms by which IKZF1 alterations impose therapy resistance on leukemic cells, including enhanced cell adhesion and modulation of glucocorticoid response.
Collapse
Affiliation(s)
- René Marke
- Laboratory of Pediatric Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Blanca Scheijen
- Laboratory of Pediatric Oncology, Radboud University Medical Center, Nijmegen, the Netherlands .,Department of Pathology, Radboud University Medical Center; Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, the Netherlands
| |
Collapse
|
29
|
Stanulla M, Dagdan E, Zaliova M, Möricke A, Palmi C, Cazzaniga G, Eckert C, Te Kronnie G, Bourquin JP, Bornhauser B, Koehler R, Bartram CR, Ludwig WD, Bleckmann K, Groeneveld-Krentz S, Schewe D, Junk SV, Hinze L, Klein N, Kratz CP, Biondi A, Borkhardt A, Kulozik A, Muckenthaler MU, Basso G, Valsecchi MG, Izraeli S, Petersen BS, Franke A, Dörge P, Steinemann D, Haas OA, Panzer-Grümayer R, Cavé H, Houlston RS, Cario G, Schrappe M, Zimmermann M. IKZF1 plus Defines a New Minimal Residual Disease-Dependent Very-Poor Prognostic Profile in Pediatric B-Cell Precursor Acute Lymphoblastic Leukemia. J Clin Oncol 2018; 36:1240-1249. [PMID: 29498923 DOI: 10.1200/jco.2017.74.3617] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Purpose Somatic deletions that affect the lymphoid transcription factor-coding gene IKZF1 have previously been reported as independently associated with a poor prognosis in pediatric B-cell precursor (BCP) acute lymphoblastic leukemia (ALL). We have now refined the prognostic strength of IKZF1 deletions by analyzing the effect of co-occurring deletions. Patients and Methods The analysis involved 991 patients with BCP ALL treated in the Associazione Italiana Ematologia ed Oncologia Pediatrica-Berlin-Frankfurt-Muenster (AIEOP-BFM) ALL 2000 trial with complete information for copy number alterations of IKZF1, PAX5, ETV6, RB1, BTG1, EBF1, CDKN2A, CDKN2B, Xp22.33/Yp11.31 (PAR1 region; CRLF2, CSF2RA, and IL3RA), and ERG; replication of findings involved 417 patients from the same trial. Results IKZF1 deletions that co-occurred with deletions in CDKN2A, CDKN2B, PAX5, or PAR1 in the absence of ERG deletion conferred the worst outcome and, consequently, were grouped as IKZF1plus. The IKZF1plus group comprised 6% of patients with BCP ALL, with a 5-year event-free survival of 53 ± 6% compared with 79 ± 5% in patients with IKZF1 deletion who did not fulfill the IKZF1plus definition and 87 ± 1% in patients who lacked an IKZF1 deletion ( P ≤ .001). Respective 5-year cumulative relapse incidence rates were 44 ± 6%, 11 ± 4%, and 10 ± 1% ( P ≤ .001). Results were confirmed in the replication cohort, and multivariable analyses demonstrated independence of IKZF1plus. The IKZF1plus prognostic effect differed dramatically in analyses stratified by minimal residual disease (MRD) levels after induction treatment: 5-year event-free survival for MRD standard-risk IKZF1plus patients was 94 ± 5% versus 40 ± 10% in MRD intermediate- and 30 ± 14% in high-risk IKZF1plus patients ( P ≤ .001). Corresponding 5-year cumulative incidence of relapse rates were 6 ± 6%, 60 ± 10%, and 60 ± 17% ( P ≤ .001). Conclusion IKZF1plus describes a new MRD-dependent very-poor prognostic profile in BCP ALL. Because current AIEOP-BFM treatment is largely ineffective for MRD-positive IKZF1plus patients, new experimental treatment approaches will be evaluated in our upcoming trial AIEOP-BFM ALL 2017.
Collapse
Affiliation(s)
- Martin Stanulla
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Elif Dagdan
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Marketa Zaliova
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Anja Möricke
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Chiara Palmi
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Giovanni Cazzaniga
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Cornelia Eckert
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Geertruy Te Kronnie
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Jean-Pierre Bourquin
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Beat Bornhauser
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Rolf Koehler
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Claus R Bartram
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Wolf-Dieter Ludwig
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Kirsten Bleckmann
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Stefanie Groeneveld-Krentz
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Denis Schewe
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Stefanie V Junk
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Laura Hinze
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Norman Klein
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Christian P Kratz
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Andrea Biondi
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Arndt Borkhardt
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Andreas Kulozik
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Martina U Muckenthaler
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Giuseppe Basso
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Maria Grazia Valsecchi
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Shai Izraeli
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Britt-Sabina Petersen
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Andre Franke
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Petra Dörge
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Doris Steinemann
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Oskar A Haas
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Renate Panzer-Grümayer
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Hélène Cavé
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Richard S Houlston
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Gunnar Cario
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Martin Schrappe
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | - Martin Zimmermann
- Martin Stanulla, Elif Dagdan, Stefanie V. Junk, Laura Hinze, Norman Klein, Christian P. Kratz, Petra Dörge, Doris Steinemann, and Martin Zimmermann, Hannover Medical School; Petra Dörge, German Center for Infection Research, Hannover; Anja Möricke, Kirsten Bleckmann, Denis Schewe, Gunnar Cario, and Martin Schrappe, University Hospital Schleswig-Holstein; Britt-Sabina Petersen and Andre Franke, Kiel University, Kiel; Cornelia Eckert and Stefanie Groeneveld-Krentz, Charité University Hospital; Wolf-Dieter Ludwig, HELIOS-Clinic Berlin-Buch, Berlin; Rolf Koehler, Claus R. Bartram, Andreas Kulozik, and Martina U. Muckenthaler, University of Heidelberg, Heidelberg; Arndt Borkhardt, Heinrich-Heine University, Düsseldorf, Germany; Marketa Zaliova, Charles University and University Hospital Motol, Prague, Czech Republic; Chiara Palmi, Giovanni Cazzaniga, and Andrea Biondi, Azienda Ospedaliera San Gerardo; Maria Grazia Valsecchi, University of Milano-Bicocca, Monza; Geertruy te Kronnie and Giuseppe Basso, University of Padova, Padua, Italy; Jean-Pierre Bourquin and Beat Bornhauser, University Children's Hospital Zurich, Zurich, Switzerland; Shai Izraeli, Sheba Medical Center Tel-Hashomer and Tel Aviv University, Tel Aviv, Israel; Oskar A. Haas and Renate Panzer-Grümayer, St Anna Kinderkrebsforschung and Medical University Vienna, Vienna, Austria; Hélène Cavé, Robert Debré Hospital and Paris-Diderot University, Paris, France; and Richard S. Houlston, The Institute of Cancer Research, London, United Kingdom
| | | | | |
Collapse
|
30
|
Genomic CDKN2A/2B deletions in adult Ph + ALL are adverse despite allogeneic stem cell transplantation. Blood 2018; 131:1464-1475. [PMID: 29348129 DOI: 10.1182/blood-2017-07-796862] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 01/09/2018] [Indexed: 12/12/2022] Open
Abstract
We investigated the role of copy number alterations to refine risk stratification in adult Philadelphia chromosome positive (Ph)+ acute lymphoblastic leukemia (ALL) treated with tyrosine kinase inhibitors (TKIs) and allogeneic stem cell transplantation (aSCT). Ninety-seven Ph+ ALL patients (median age 41 years; range 18-64 years) within the prospective multicenter German Multicenter ALL Study Group studies 06/99 (n = 8) and 07/2003 (n = 89) were analyzed. All patients received TKI and aSCT in first complete remission (CR1). Copy number analysis was performed with single nucleotide polymorphism arrays and validated by multiplex ligation-dependent probe amplification. The frequencies of recurrently deleted genes were: IKZF1, 76%; CDKN2A/2B, 45%; PAX5, 43%; BTG1, 18%; EBF1, 13%; ETV6, 5%; RB, 14%. In univariate analyses, the presence of CDKN2A/2B deletions had a negative impact on all endpoints: overall survival (P = .023), disease-free survival (P = .012), and remission duration (P = .036). The negative predictive value of CDKN2A/2B deletions was retained in multivariable analysis along with other factors such as timing of TKI therapy, intensity of conditioning, achieving remission after induction phase 1 and BTG1 deletions. We therefore conclude that acquired genomic CDKN2A/2B deletions identify a subgroup of Ph+ ALL patients, who have an inferior prognosis despite aSCT in CR1. Their poor outcome was attributable primarily to a high relapse rate after aSCT.
Collapse
|
31
|
Sutton R, Venn NC, Law T, Boer JM, Trahair TN, Ng A, Den Boer ML, Dissanayake A, Giles JE, Dalzell P, Mayoh C, Barbaric D, Revesz T, Alvaro F, Pieters R, Haber M, Norris MD, Schrappe M, Dalla Pozza L, Marshall GM. A risk score including microdeletions improves relapse prediction for standard and medium risk precursor B-cell acute lymphoblastic leukaemia in children. Br J Haematol 2017; 180:550-562. [DOI: 10.1111/bjh.15056] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Rosemary Sutton
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
- School of Women's and Children's Health; UNSW; Sydney Australia
| | - Nicola C. Venn
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
| | - Tamara Law
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
| | - Judith M. Boer
- Department of Paediatric Oncology/Haematology; Erasmus Medical Centre; Sophia Children's Hospital; Rotterdam The Netherlands
- Princess Máxima Centre for Paediatric Oncology; Utrecht The Netherlands
| | - Toby N. Trahair
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
- School of Women's and Children's Health; UNSW; Sydney Australia
- Kids Cancer Centre; Sydney Children's Hospital; Randwick Australia
| | - Anthea Ng
- Cancer Centre for Children; The Children's Hospital at Westmead; Westmead Australia
| | - Monique L. Den Boer
- Department of Paediatric Oncology/Haematology; Erasmus Medical Centre; Sophia Children's Hospital; Rotterdam The Netherlands
- Dutch Childhood Oncology Group; The Hague The Netherlands
| | | | - Jodie E. Giles
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
| | | | - Chelsea Mayoh
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
| | - Draga Barbaric
- Kids Cancer Centre; Sydney Children's Hospital; Randwick Australia
| | - Tamas Revesz
- Women's and Children's Hospital, SA Pathology; University of Adelaide; Adelaide Australia
| | - Frank Alvaro
- John Hunter Children's Hospital; Newcastle Australia
| | - Rob Pieters
- Princess Máxima Centre for Paediatric Oncology; Utrecht The Netherlands
- Dutch Childhood Oncology Group; The Hague The Netherlands
| | - Michelle Haber
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
- School of Women's and Children's Health; UNSW; Sydney Australia
| | - Murray D. Norris
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
- UNSW Centre for Childhood Cancer Research; Kensington Australia
| | - Martin Schrappe
- Christian-Albrechts-University Kiel and University Medical Centre Schleswig-Holstein; Kiel Germany
| | - Luciano Dalla Pozza
- Cancer Centre for Children; The Children's Hospital at Westmead; Westmead Australia
| | - Glenn M Marshall
- Children's Cancer Institute; Lowy Cancer Research Centre; UNSW; Sydney Australia
- School of Women's and Children's Health; UNSW; Sydney Australia
- Kids Cancer Centre; Sydney Children's Hospital; Randwick Australia
| |
Collapse
|
32
|
Arber DA, Borowitz MJ, Cessna M, Etzell J, Foucar K, Hasserjian RP, Rizzo JD, Theil K, Wang SA, Smith AT, Rumble RB, Thomas NE, Vardiman JW. Initial Diagnostic Workup of Acute Leukemia: Guideline From the College of American Pathologists and the American Society of Hematology. Arch Pathol Lab Med 2017; 141:1342-1393. [PMID: 28225303 DOI: 10.5858/arpa.2016-0504-cp] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - A complete diagnosis of acute leukemia requires knowledge of clinical information combined with morphologic evaluation, immunophenotyping and karyotype analysis, and often, molecular genetic testing. Although many aspects of the workup for acute leukemia are well accepted, few guidelines have addressed the different aspects of the diagnostic evaluation of samples from patients suspected to have acute leukemia. OBJECTIVE - To develop a guideline for treating physicians and pathologists involved in the diagnostic and prognostic evaluation of new acute leukemia samples, including acute lymphoblastic leukemia, acute myeloid leukemia, and acute leukemias of ambiguous lineage. DESIGN - The College of American Pathologists and the American Society of Hematology convened a panel of experts in hematology and hematopathology to develop recommendations. A systematic evidence review was conducted to address 6 key questions. Recommendations were derived from strength of evidence, feedback received during the public comment period, and expert panel consensus. RESULTS - Twenty-seven guideline statements were established, which ranged from recommendations on what clinical and laboratory information should be available as part of the diagnostic and prognostic evaluation of acute leukemia samples to what types of testing should be performed routinely, with recommendations on where such testing should be performed and how the results should be reported. CONCLUSIONS - The guideline provides a framework for the multiple steps, including laboratory testing, in the evaluation of acute leukemia samples. Some aspects of the guideline, especially molecular genetic testing in acute leukemia, are rapidly changing with new supportive literature, which will require on-going updates for the guideline to remain relevant.
Collapse
|
33
|
Pan L, Liu G, Lin F, Zhong S, Xia H, Sun X, Liang H. Machine learning applications for prediction of relapse in childhood acute lymphoblastic leukemia. Sci Rep 2017; 7:7402. [PMID: 28784991 PMCID: PMC5547099 DOI: 10.1038/s41598-017-07408-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/23/2017] [Indexed: 01/17/2023] Open
Abstract
The prediction of relapse in childhood acute lymphoblastic leukemia (ALL) is a critical factor for successful treatment and follow-up planning. Our goal was to construct an ALL relapse prediction model based on machine learning algorithms. Monte Carlo cross-validation nested by 10-fold cross-validation was used to rank clinical variables on the randomly split training sets of 336 newly diagnosed ALL children, and a forward feature selection algorithm was employed to find the shortest list of most discriminatory variables. To enable an unbiased estimation of the prediction model to new patients, besides the split test sets of 150 patients, we introduced another independent data set of 84 patients to evaluate the model. The Random Forest model with 14 features achieved a cross-validation accuracy of 0.827 ± 0.031 on one set and an accuracy of 0.798 on the other, with the area under the curve of 0.902 ± 0.027 and 0.904, respectively. The model performed well across different risk-level groups, with the best accuracy of 0.829 in the standard-risk group. To our knowledge, this is the first study to use machine learning models to predict childhood ALL relapse based on medical data from Electronic Medical Record, which will further facilitate stratification treatments.
Collapse
Affiliation(s)
- Liyan Pan
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Guangjian Liu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Fangqin Lin
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shuling Zhong
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xin Sun
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Huiying Liang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
34
|
Kobitzsch B, Gökbuget N, Schwartz S, Reinhardt R, Brüggemann M, Viardot A, Wäsch R, Starck M, Thiel E, Hoelzer D, Burmeister T. Loss-of-function but not dominant-negative intragenic IKZF1 deletions are associated with an adverse prognosis in adult BCR-ABL-negative acute lymphoblastic leukemia. Haematologica 2017; 102:1739-1747. [PMID: 28751559 PMCID: PMC5622858 DOI: 10.3324/haematol.2016.161273] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/18/2017] [Indexed: 12/17/2022] Open
Abstract
Genetic alterations of the transcription factor IKZF1 ("IKAROS") are detected in around 15-30% of cases of BCR-ABL-negative B-cell precursor acute lymphoblastic leukemia. Different types of intragenic deletions have been observed, resulting in a functionally inactivated allele ("loss-of-function") or in "dominant-negative" isoforms. The prognostic impact of these alterations especially in adult acute lymphoblastic leukemia is not well defined. We analyzed 482 well-characterized cases of adult BCR-ABL-negative B-precursor acute lymphoblastic leukemia uniformly treated in the framework of the GMALL studies and detected IKZF1 alterations in 128 cases (27%). In 20%, the IKZF1 alteration was present in a large fraction of leukemic cells ("high deletion load") while in 7% it was detected only in small subclones ("low deletion load"). Some patients showed more than one IKZF1 alteration (8%). Patients exhibiting a loss-of-function isoform with high deletion load had a shorter overall survival (OS at 5 years 28% vs. 59%; P<0.0001), also significant in a subgroup analysis of standard risk patients according to GMALL classification (OS at 5 years 37% vs. 68%; P=0.0002). Low deletion load or dominant-negative IKZF1 alterations had no prognostic impact. The results thus suggest that there is a clear distinction between loss-of-function and dominant-negative IKZF1 deletions. Affected patients should thus be monitored for minimal residual disease carefully to detect incipient relapses at an early stage and they are potential candidates for alternative or intensified treatment regimes. (clinicaltrials.gov identifiers: 00199056 and 00198991).
Collapse
Affiliation(s)
- Benjamin Kobitzsch
- Department of Hematology, Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Germany
| | - Nicola Gökbuget
- Department of Medicine II, Hematology/Oncology, Goethe University, Frankfurt/Main, Germany
| | - Stefan Schwartz
- Department of Hematology, Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Germany
| | | | - Monika Brüggemann
- Department of Hematology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Andreas Viardot
- Department of Medicine III (Hematology, Oncology), Ulm University, Munich, Germany
| | - Ralph Wäsch
- Department of Hematology, Oncology and Stem Cell Transplantation, University of Freiburg Medical Center, Munich, Germany
| | - Michael Starck
- Department of Hematology, Klinikum München-Schwabing, Munich, Germany
| | - Eckhard Thiel
- Department of Hematology, Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Germany
| | - Dieter Hoelzer
- Department of Medicine II, Hematology/Oncology, Goethe University, Frankfurt/Main, Germany
| | - Thomas Burmeister
- Department of Hematology, Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Germany
| |
Collapse
|
35
|
Abstract
Acute lymphoblastic leukemia (ALL) is characterized by a great biological and clinical heterogeneity. Despite most adult patients enter complete hematologic remission after induction therapy only 40% survive five or more years. Over the last 20 years, the definition of an accurate biologic leukemia profile and the minimal residual disease evaluation in addition to conventional risk criteria led to a significant improvement for the risk stratification. The alterations of the oncosuppressor gene TP53, including deletions, sequence mutations and defect in its expression due to regulatory defects, define a new important predictor of adverse outcome. More recently, new drugs have been developed with the aim of targeting p53 protein itself or its regulatory molecules, such as Mdm2, and restoring the pathway functionality. Therefore, TP53 alterations should be considered in the diagnostic work-up to identify high risk ALL patients in need of intensive treatment strategies or eligible for new innovative targeted therapies.
Collapse
Affiliation(s)
- Silvia Salmoiraghi
- a Hematology and Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII , Bergamo , Italy
| | - Alessandro Rambaldi
- a Hematology and Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII , Bergamo , Italy.,b Department of Hematology-Oncology , University of Milan , Milan , Italy
| | - Orietta Spinelli
- a Hematology and Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII , Bergamo , Italy
| |
Collapse
|
36
|
Bhandari P, Ahmad F, Das BR. Molecular profiling of gene copy number abnormalities in key regulatory genes in high-risk B-lineage acute lymphoblastic leukemia: frequency and their association with clinicopathological findings in Indian patients. Med Oncol 2017; 34:92. [PMID: 28401483 DOI: 10.1007/s12032-017-0940-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023]
Abstract
Genes related to key cellular pathways are frequently altered in B cell ALL and are associated with poor survival especially in high-risk (HR) subgroups. We examined gene copy number abnormalities (CNA) in 101 Indian HR B cell ALL patients and their correlation with clinicopathological features by multiplex ligation-dependent probe amplification. Overall, CNA were detected in 59 (59%) cases, with 26, 10 and 23% of cases harboring 1, 2 or +3 CNA. CNA were more prevalent in BCR-ABL1 (60%), pediatric (64%) and high WCC (WBC count) (63%) patients. Frequent genes deletions included CDNK2A/B (26%), IKZF1 (25%), PAX5 (14%), JAK2 (7%), BTG1 (6%), RB1 (5%), EBF1 (4%), ETV6 (4%), while PAR1 region genes were predominantly duplicated (20%). EBF1 deletions selectively associated with adults, IKZF1 deletions occurred frequently in high WCC and BCR-ABL1 cases, while PAR1 region gains significantly associated with MLL-AF4 cases. IKZF1 haploinsufficiency group was predominant, especially in adults (65%), high WCC (60%) patients and BCR-ABL1-negative (78%) patients. Most cases harbored multiple concurrent CNA, with IKZF1 concomitantly occurring with CDNK2A/B, PAX5 and BTG1, while JAK2 occurred with CDNK2A/B and PAX5. Mutually exclusive CNA included ETV6 and IKZF1/RB1, and EBF1 and JAK2. Our results corroborate with global reports, aggregating molecular markers in Indian HR B-ALL cases. Integration of CNA data from rapid methods like MLPA, onto background of existing gold-standard methods detecting significant chromosomal abnormalities, provides a comprehensive genetic profile in B-ALL.
Collapse
Affiliation(s)
- Prerana Bhandari
- Research and Development Division, Molecular Pathology, Clinical Research Services, SRL Limited, Plot No.1, Prime Square Building, S.V. Road, Goregaon (W), Mumbai, 400062, India
| | - Firoz Ahmad
- Research and Development Division, Molecular Pathology, Clinical Research Services, SRL Limited, Plot No.1, Prime Square Building, S.V. Road, Goregaon (W), Mumbai, 400062, India
| | - Bibhu Ranjan Das
- Research and Development Division, Molecular Pathology, Clinical Research Services, SRL Limited, Plot No.1, Prime Square Building, S.V. Road, Goregaon (W), Mumbai, 400062, India.
| |
Collapse
|
37
|
Witkowski MT, Hu Y, Roberts KG, Boer JM, McKenzie MD, Liu GJ, Le Grice OD, Tremblay CS, Ghisi M, Willson TA, Horstmann MA, Aifantis I, Cimmino L, Frietze S, den Boer ML, Mullighan CG, Smyth GK, Dickins RA. Conserved IKAROS-regulated genes associated with B-progenitor acute lymphoblastic leukemia outcome. J Exp Med 2017; 214:773-791. [PMID: 28190000 PMCID: PMC5339666 DOI: 10.1084/jem.20160048] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 10/05/2016] [Accepted: 12/19/2016] [Indexed: 12/31/2022] Open
Abstract
Genetic alterations disrupting the transcription factor IKZF1 (encoding IKAROS) are associated with poor outcome in B lineage acute lymphoblastic leukemia (B-ALL) and occur in >70% of the high-risk BCR-ABL1+ (Ph+) and Ph-like disease subtypes. To examine IKAROS function in this context, we have developed novel mouse models allowing reversible RNAi-based control of Ikaros expression in established B-ALL in vivo. Notably, leukemias driven by combined BCR-ABL1 expression and Ikaros suppression rapidly regress when endogenous Ikaros is restored, causing sustained disease remission or ablation. Comparison of transcriptional profiles accompanying dynamic Ikaros perturbation in murine B-ALL in vivo with two independent human B-ALL cohorts identified nine evolutionarily conserved IKAROS-repressed genes. Notably, high expression of six of these genes is associated with inferior event-free survival in both patient cohorts. Among them are EMP1, which was recently implicated in B-ALL proliferation and prednisolone resistance, and the novel target CTNND1, encoding P120-catenin. We demonstrate that elevated Ctnnd1 expression contributes to maintenance of murine B-ALL cells with compromised Ikaros function. These results suggest that IKZF1 alterations in B-ALL leads to induction of multiple genes associated with proliferation and treatment resistance, identifying potential new therapeutic targets for high-risk disease.
Collapse
Affiliation(s)
- Matthew T Witkowski
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia.,Molecular Medicine Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville 3010, Victoria, Australia
| | - Yifang Hu
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
| | - Kathryn G Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Judith M Boer
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, 3015 CN Rotterdam, Netherlands
| | - Mark D McKenzie
- Molecular Medicine Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville 3010, Victoria, Australia
| | - Grace J Liu
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia.,Molecular Medicine Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville 3010, Victoria, Australia
| | - Oliver D Le Grice
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia.,Molecular Medicine Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
| | - Cedric S Tremblay
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - Margherita Ghisi
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - Tracy A Willson
- Molecular Medicine Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
| | - Martin A Horstmann
- Research Institute Children's Cancer Center, Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, 20246 Hamburg, Germany
| | - Iannis Aifantis
- Department of Pathology, NYU School of Medicine, New York, NY 10016
| | - Luisa Cimmino
- Department of Pathology, NYU School of Medicine, New York, NY 10016
| | - Seth Frietze
- Department of Medical Laboratory and Radiation Science, University of Vermont, Burlington, VT 05405
| | - Monique L den Boer
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, 3015 CN Rotterdam, Netherlands.,Dutch Childhood Oncology Group, 2545 The Hague, Netherlands
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Gordon K Smyth
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.,Department of Mathematics and Statistics, University of Melbourne, Parkville 3010, Victoria, Australia
| | - Ross A Dickins
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia .,Molecular Medicine Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville 3010, Victoria, Australia
| |
Collapse
|
38
|
Short NJ, Jabbour E. Minimal Residual Disease in Acute Lymphoblastic Leukemia: How to Recognize and Treat It. Curr Oncol Rep 2017; 19:6. [DOI: 10.1007/s11912-017-0565-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
39
|
Scheijen B, Boer JM, Marke R, Tijchon E, van Ingen Schenau D, Waanders E, van Emst L, van der Meer LT, Pieters R, Escherich G, Horstmann MA, Sonneveld E, Venn N, Sutton R, Dalla-Pozza L, Kuiper RP, Hoogerbrugge PM, den Boer ML, van Leeuwen FN. Tumor suppressors BTG1 and IKZF1 cooperate during mouse leukemia development and increase relapse risk in B-cell precursor acute lymphoblastic leukemia patients. Haematologica 2016; 102:541-551. [PMID: 27979924 PMCID: PMC5394950 DOI: 10.3324/haematol.2016.153023] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/14/2016] [Indexed: 12/16/2022] Open
Abstract
Deletions and mutations affecting lymphoid transcription factor IKZF1 (IKAROS) are associated with an increased relapse risk and poor outcome in B-cell precursor acute lymphoblastic leukemia. However, additional genetic events may either enhance or negate the effects of IKZF1 deletions on prognosis. In a large discovery cohort of 533 childhood B-cell precursor acute lymphoblastic leukemia patients, we observed that single-copy losses of BTG1 were significantly enriched in IKZF1-deleted B-cell precursor acute lymphoblastic leukemia (P=0.007). While BTG1 deletions alone had no impact on prognosis, the combined presence of BTG1 and IKZF1 deletions was associated with a significantly lower 5-year event-free survival (P=0.0003) and a higher 5-year cumulative incidence of relapse (P=0.005), when compared with IKZF1-deleted cases without BTG1 aberrations. In contrast, other copy number losses commonly observed in B-cell precursor acute lymphoblastic leukemia, such as CDKN2A/B, PAX5, EBF1 or RB1, did not affect the outcome of IKZF1-deleted acute lymphoblastic leukemia patients. To establish whether the combined loss of IKZF1 and BTG1 function cooperate in leukemogenesis, Btg1-deficient mice were crossed onto an Ikzf1 heterozygous background. We observed that loss of Btg1 increased the tumor incidence of Ikzf1+/− mice in a dose-dependent manner. Moreover, murine B cells deficient for Btg1 and Ikzf1+/− displayed increased resistance to glucocorticoids, but not to other chemotherapeutic drugs. Together, our results identify BTG1 as a tumor suppressor in leukemia that, when deleted, strongly enhances the risk of relapse in IKZF1-deleted B-cell precursor acute lymphoblastic leukemia, and augments the glucocorticoid resistance phenotype mediated by the loss of IKZF1 function.
Collapse
Affiliation(s)
- Blanca Scheijen
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, the Netherlands
| | - Judith M Boer
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - René Marke
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, the Netherlands
| | - Esther Tijchon
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, the Netherlands
| | | | - Esmé Waanders
- Department of Human Genetics, Radboud university medical center, Nijmegen, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Liesbeth van Emst
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, the Netherlands
| | - Laurens T van der Meer
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, the Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Gabriele Escherich
- Research Institute Children's Cancer Center and Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin A Horstmann
- Research Institute Children's Cancer Center and Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Nicola Venn
- Australian and New Zealand Children's Oncology Group, Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Rosemary Sutton
- Australian and New Zealand Children's Oncology Group, Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | | | - Roland P Kuiper
- Department of Human Genetics, Radboud university medical center, Nijmegen, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Monique L den Boer
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, the Netherlands
| |
Collapse
|
40
|
Zhao S, Liu W, Li Y, Liu P, Li S, Dou D, Wang Y, Yang R, Xiang R, Liu F. Alternative Splice Variants Modulates Dominant-Negative Function of Helios in T-Cell Leukemia. PLoS One 2016; 11:e0163328. [PMID: 27681508 PMCID: PMC5040427 DOI: 10.1371/journal.pone.0163328] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/06/2016] [Indexed: 12/21/2022] Open
Abstract
The molecular defects which lead to multistep incidences of human T-cell leukemia have yet to be identified. The DNA-binding protein Helios (known as IKZF2), a member of the Ikaros family of Krüppel-like zinc-finger proteins, functions pivotally in T-cell differentiation and activation. In this study, we identify three novel short Helios splice variants which are T-cell leukemic specific, and demonstrate their dominant-negative function. We then test the cellular localization of distinct Helios isoforms, as well as their capability to form heterodimer with Ikaros, and the association with complexes comprising histone deacetylase (HDAC). In addition, the ectopic expression of T-cell leukemic Helios isoforms interferes with T-cell proliferation and apoptosis. The gene expression profiling and pathway analysis indicated the enrichment of signaling pathways essential for gene expression, translation, cell cycle checkpoint, and response to DNA damage stimulus. These data indicate the molecular function of Helios to be involved in the leukemogenesis and phenotype of T-cell leukemia, and also reveal Helios deregulation as a novel marker for T-cell leukemia.
Collapse
Affiliation(s)
- Shaorong Zhao
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Wei Liu
- Tianjin Entry-Exit Inspection and Quarantine Bureau, Tianjin 300308, China
| | - Yinghui Li
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Pengjiang Liu
- Department of Hematology, First-Central Hospital, Tianjin 300060, China
| | - Shufang Li
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Daolei Dou
- State Key Laboratory of Medical Chemical Biology, Tianjin 300070, China
| | - Yue Wang
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Rongcun Yang
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Rong Xiang
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Tianjin 300071, China
- * E-mail: (FL); (RX)
| | - Feifei Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
- * E-mail: (FL); (RX)
| |
Collapse
|
41
|
Besbes S, Hamadou WS, Boulland ML, Lefranc MP, Ben Youssef Y, Achour B, Khelif A, Fest T, Soua Z. Combined IKZF1 and IG markers as new tools for diagnosis and minimal residual disease assessment in Tunisian B-ALL. Bull Cancer 2016; 103:822-828. [PMID: 27614734 DOI: 10.1016/j.bulcan.2016.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/21/2016] [Accepted: 07/23/2016] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The monitoring of minimal residual disease (MRD) approach in patients diagnosed with B-acute lymphoblastic leukemia (B-ALL) allows an early detection of residual clones inducing relapses and therefore appropriate therapy strategy. The molecular markers may identify and quantify the residual blasts in B-ALL with normal cytology. In this study, we aimed to use combined IKZF1, IGH and IGK immunoglobulin genes for diagnosis and MRD monitoring in B-ALL sample using MLPA, multiplex PCR and real-time quantitative PCR. MATERIAL We showed that multiplex PCR and MLPA are necessary and complementary to detect IKZF1 deletions. RESULTS We have identified at the diagnosis clonal IGH rearrangement (VH3-JH5) and IKZF1 deletion (Δ4-7), which we have used it for MRD evaluation after induction chemotherapy. Despite the absence of chromosome abnormality, the patient may be classified in high-risk group with a relapse rate of residual blasts>10-4 and sensitivity up to 10-5. This molecular approach enabled the patient's stratification, which was overlooked by classical methods. CONCLUSION The combined IKZF1 and immunoglobulin genes will be used as appropriate molecular tools for diagnosis and MRD assessment of B-lineage leukemias and introduced as a routine tests in Tunisian clinical laboratories. They will be useful to stratify patients into risk groups leading to better treatment strategy.
Collapse
Affiliation(s)
- Sawsen Besbes
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia
| | - Walid-Sabri Hamadou
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia
| | | | | | - Yosra Ben Youssef
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia; CHU F. Hached, service d'hématologie clinique, 4000 Sousse, Tunisia
| | - Béchir Achour
- CHU F. Hached, service d'hématologie clinique, 4000 Sousse, Tunisia
| | - Abderrahim Khelif
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia; CHU F. Hached, service d'hématologie clinique, 4000 Sousse, Tunisia
| | - Thierry Fest
- CHU Pontchaillou, service d'hématologie biologique, 35033 Rennes, France
| | - Zohra Soua
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia.
| |
Collapse
|
42
|
Li HF, Meng WT, Jia YQ, Jiang NG, Zeng TT, Jin YM, Huang QR, Li X, Xu H, Mo XM. Development-associated immunophenotypes reveal the heterogeneous and individualized early responses of adult B-acute lymphoblastic leukemia. Medicine (Baltimore) 2016; 95:e4128. [PMID: 27559941 PMCID: PMC5400307 DOI: 10.1097/md.0000000000004128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
B cell acute lymphoblastic leukemia (B-ALL) exhibits phenotypes reminiscent of normal stages of B-cell development. As demonstrated by flow cytometry, the immunophenotypes are able to determine the stages of B cell development. Multicolor flow cytometry (MFC) is more accurate at identifying cell populations. In this study, 9-color panels, including CD10, CD19, CD20, CD22, CD34, CD79a, CD179a, and IgM, which are sequentially expressed during B cell development, were designed to detect the leukemia cell subpopulations in adult B-ALL patients. In 23 patients at diagnosis, 192 heterogeneous subpopulations of leukemia cells were detected. Compared with their counterparts at diagnosis and after the 1st course of induction therapy, the responses of the subpopulations were also heterogeneous. In the CD10 population, the residual B cell subpopulations in the BCR/ABL patients were obviously reduced compared to those in the BCR/ABL patients. New subpopulations were detected in 22 of 23 patients and were primarily located in the CD34CD10 populations. Subpopulations of clonal evolution were heterogeneous after induction therapy. Our results suggest that the subpopulations in B-ALL patients should be dynamically monitored by development-associated immunophenotyping before, during, and after induction therapy and to predict the prognosis of the disease.
Collapse
Affiliation(s)
- Hui-Fang Li
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Wen-Tong Meng
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
- Correspondence: Wen-Tong Meng, Xian-Ming Mo, Collaborative Innovation Center for Biotherapy, Sichuan University, Building B2, No. 88, Keyuan Nanlu, High-Tech District, Chengdu 610041, China (e-mail: , )
| | | | | | | | - Yong-Mei Jin
- Department of Laboratory Medicine, West China Hospital
| | - Qiao-Rong Huang
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xue Li
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Hong Xu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xian-Ming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
- Correspondence: Wen-Tong Meng, Xian-Ming Mo, Collaborative Innovation Center for Biotherapy, Sichuan University, Building B2, No. 88, Keyuan Nanlu, High-Tech District, Chengdu 610041, China (e-mail: , )
| |
Collapse
|
43
|
Paietta E. Minimal Residual Disease in AML: Why Has It Lagged Behind Pediatric ALL? CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2016; 15 Suppl:S2-6. [PMID: 26297274 DOI: 10.1016/j.clml.2015.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 02/03/2015] [Indexed: 12/16/2022]
Abstract
Although the concept of minimal residual disease (MRD) as an indicator for the quality of treatment response is the same in acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL), the practice of measuring MRD levels for monitoring response and guiding therapy after induction has been implemented much more rapidly in ALL, particularly pediatric ALL, than in AML. In this perspective we examine the facts and discuss why ALL appears to be more amenable to MRD-shaped risk allocation and a revised definition of complete remission.
Collapse
Affiliation(s)
- Elisabeth Paietta
- Montefiore Medical Center-North Division, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
44
|
IKZF1 deletion is enriched in pediatric B-cell precursor acute lymphoblastic leukemia patients showing prednisolone resistance. Leukemia 2016; 30:1801-3. [PMID: 27198053 DOI: 10.1038/leu.2016.128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Moorman AV. New and emerging prognostic and predictive genetic biomarkers in B-cell precursor acute lymphoblastic leukemia. Haematologica 2016; 101:407-16. [PMID: 27033238 PMCID: PMC5004393 DOI: 10.3324/haematol.2015.141101] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/19/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a heterogeneous disease at the genetic level. Chromosomal abnormalities are used as diagnostic, prognostic and predictive biomarkers to provide subtype, outcome and drug response information. t(12;21)/ETV6-RUNX1 and high hyper-diploidy are good-risk prognostic biomarkers whereas KMT2A(MLL) translocations, t(17;19)/TCF3-HLF, haploidy or low hypodiploidy are high-risk biomarkers. t(9;22)/BCR-ABL1 patients require targeted treatment (imatinib/dasatinib), whereas iAMP21 patients achieve better outcomes when treated intensively. High-risk genetic biomarkers are four times more prevalent in adults compared to children. The application of genomic technologies to cases without an established abnormality (B-other) reveals copy number alterations which can be used either individually or in combination as prognostic biomarkers. Transcriptome sequencing studies have identified a network of fusion genes involving kinase genes -ABL1,ABL2,PDGFRB,CSF1R,CRLF2,JAK2 and EPOR in-vitro and in-vivo studies along with emerging clinical observations indicate that patients with a kinase-activating aberration may respond to treatment with small molecular inhibitors like imatinib/dasatinib and ruxolitinib. Further work is required to determine the true frequency of these abnormalities across the age spectrum and the optimal way to incorporate such inhibitors into protocols. In conclusion, genetic biomarkers are playing an increasingly important role in the management of patients with ALL.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- B-Lymphocytes/drug effects
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Chromosomes, Human, Pair 12
- Chromosomes, Human, Pair 21
- Chromosomes, Human, Pair 22
- Chromosomes, Human, Pair 9
- Dasatinib/therapeutic use
- Gene Expression
- Humans
- Imatinib Mesylate/therapeutic use
- Nitriles
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality
- Prognosis
- Pyrazoles/therapeutic use
- Pyrimidines
- Survival Analysis
- Translocation, Genetic
Collapse
Affiliation(s)
- Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
46
|
Abstract
The prognostic power of minimal residual disease after therapy for acute leukemias is not in question. It is only logical that the finding of leukemic blast cells after therapy predicts for impending relapse or at least the need for additional treatment. Which level of what is called minimal residual disease (MRD) is clinically relevant, however, depends on the efficacy of the initial treatment as well as the treatment strategies available to target MRD. There are a multitude of additional factors that can alter the clinical significance of MRD, including the genotype of the patient's leukemic cells. The fact that methodologies of MRD detection are not standardized and thresholds for defining MRD positivity vary depending upon MRD detection method and the operator's skills or convictions only add to the complexity of MRD interpretation. While enormous efforts are devoted to enhancing the sensitivity of MRD detection, eg, by next-generation sequencing, improvements of methods for detecting MRD per se will not automatically lead to a more reliable estimation of total tumor burden. Most importantly, even the best assay will yield accurate MRD results only if the tissue source for MRD determination is of good quality. Another aspect of potentially crucial importance is the heterogenous distribution of leukemic cells throughout the skeleton after treatment, recently demonstrated for acute myeloid leukemia (AML) by bone marrow imaging. Once technical difficulties of MRD measurement are resolved and better MRD-targeting drugs are developed, we still need to learn about alternate proposed mechanisms to explain MRD-independent prognostication, well described in acute lymphoid leukemia, before MRD can be included routinely in the guidance of therapy in AML.
Collapse
Affiliation(s)
- Elisabeth Paietta
- Montefiore Medical Center, Albert Einstein College of Medicine, 111East 210th Street, Bronx, NY 10467, USA.
| |
Collapse
|
47
|
Loghavi S, Kutok JL, Jorgensen JL. B-acute lymphoblastic leukemia/lymphoblastic lymphoma. Am J Clin Pathol 2015; 144:393-410. [PMID: 26276770 DOI: 10.1309/ajcpan7bh5dnywzb] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES This session of the 2013 Society of Hematopathology/European Association for Haematopathology Workshop was dedicated to B-acute lymphoblastic leukemia (B-ALL)/lymphoblastic lymphoma (LBL) with recurrent translocations and not otherwise specified. METHODS In this review, we summarize the cases discussed during the workshop, review the pertinent and most recent literature on the respective topics, and provide a few key points that may aid in the workup of patients with B-ALL/LBL. RESULTS Many of the submitted cases showed interesting diagnostic, immunophenotypic, or clinical aspects of B-ALL with BCR/ABL1, MLL-associated, and other recurrent chromosomal abnormalities. Several cases showed rare aberrancies such as coexistent IGH/BCL2 and MYC rearrangements and raised issues in classification. Other cases had unusual clinical presentations, including B-ALL with hypereosinophilia and therapy-related B-ALL. Several cases highlighted the role of flow cytometry immunophenotyping in distinguishing benign B-cell precursors from aberrant lymphoblasts, and other cases raised questions regarding the clinical importance of myeloperoxidase positivity in acute lymphoblastic leukemia. CONCLUSIONS The complexity and spectrum of cases presented in this review highlight the importance of clinicopathologic correlation and the value of ancillary studies in the classification and workup of patients with B-ALL/LBL.
Collapse
Affiliation(s)
- Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston; and
| | | | - Jeffrey L. Jorgensen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston; and
| |
Collapse
|
48
|
van der Sligte NE, Scherpen FJG, Ter Elst A, Guryev V, van Leeuwen FN, de Bont ESJM. Effect of IKZF1 deletions on signal transduction pathways in Philadelphia chromosome negative pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Exp Hematol Oncol 2015; 4:23. [PMID: 26269779 PMCID: PMC4534008 DOI: 10.1186/s40164-015-0017-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 11/13/2022] Open
Abstract
Background IKZF1 deletions are an unfavorable prognostic factor in children with Philadelphia chromosome positive (Ph+) as well as negative (Ph−) acute lymphoblastic leukemia (ALL). Although IKZF1 deletions occur in 10–15% of Ph− ALL cases, effects of IKZF1 deletions on signaling pathways in this group have not been extensively studied. Therefore, in this study we aimed to study the effect of IKZF1 deletions on active signal transduction pathways. Methods Multiplex ligation-dependent probe amplification (MLPA) was used to determine IKZF1 deletions and other copy number alterations in 109 pediatric B-Cell Precursor ALL (BCP-ALL) patients. Kinase activity profiling of 45 primary Ph− BCP-ALL patients (31 IKZF1 wild type patients and 14 patients harboring an IKZF1 alteration) and western blot analysis of 14 pediatric BCP-ALL samples was performed to determine active signal transduction pathways. Results Unsupervised hierarchical cluster analysis of kinome profiles of 45 pediatric Ph− ALL cases showed no clustering based on IKZF1 status. Comparing the phosphorylation intensities of peptides associated with signaling pathways known to be involved in BCP-ALL maintenance, we did not observe differences between the two groups. Western blot analysis of 14 pediatric BCP-ALL samples showed large variations in phosphorylation levels between the different ALL samples, independent of IKZF1 status. Conclusions Based on these results we conclude that, although IKZF1 deletions appear to be an important clinical prognostic factor, we were unable to identify a unique IKZF1 dependent protein expression signature in pediatric Ph− ALL and consequently no specific targets for future therapy of Ph−IKZF1 deleted BCP-ALL could be identified. Electronic supplementary material The online version of this article (doi:10.1186/s40164-015-0017-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Naomi E van der Sligte
- Division of Pediatric Oncology/Hematology, Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Frank J G Scherpen
- Division of Pediatric Oncology/Hematology, Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Arja Ter Elst
- Division of Pediatric Oncology/Hematology, Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eveline S J M de Bont
- Division of Pediatric Oncology/Hematology, Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
49
|
Prognostic value of rare IKZF1 deletion in childhood B-cell precursor acute lymphoblastic leukemia: an international collaborative study. Leukemia 2015. [PMID: 26202931 DOI: 10.1038/leu.2015.199] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Deletions in IKZF1 are found in ~15% of children with B-cell precursor acute lymphoblastic leukemia (BCP-ALL). There is strong evidence for the poor prognosis of IKZF1 deletions affecting exons 4-7 and exons 1-8, but evidence for the remaining 33% of cases harboring other variants of IKZF1 deletions is lacking. In an international multicenter study we analyzed the prognostic value of these rare variants in a case-control design. Each IKZF1-deleted case was matched to three IKZF1 wild-type controls based on cytogenetic subtype, treatment protocol, risk stratification arm, white blood cell count and age. Hazard ratios for the prognostic impact of rare IKZF1 deletions on event-free survival were calculated by matched pair Cox regression. Matched pair analysis for all 134 cases with rare IKZF1 deletions together revealed a poor prognosis (P<0.001) that was evident in each risk stratification arm. Rare variant types with the most unfavorable event-free survival were DEL 2-7 (P=0.03), DEL 2-8 (P=0.002) and DEL-Other (P<0.001). The prognosis of each type of rare variant was equal or worse compared with the well-known major DEL 4-7 and DEL 1-8 IKZF1 deletion variants. We therefore conclude that all variants of rare IKZF1 deletions are associated with an unfavorable prognosis in pediatric BCP-ALL.
Collapse
|
50
|
Ghazavi F, Clappier E, Lammens T, Suciu S, Caye A, Zegrari S, Bakkus M, Grardel N, Benoit Y, Bertrand Y, Minckes O, Costa V, Ferster A, Mazingue F, Plat G, Plouvier E, Poirée M, Uyttebroeck A, van der Werff-Ten Bosch J, Yakouben K, Helsmoortel H, Meul M, Van Roy N, Philippé J, Speleman F, Cavé H, Van Vlierberghe P, De Moerloose B. CD200/BTLA deletions in pediatric precursor B-cell acute lymphoblastic leukemia treated according to the EORTC-CLG 58951 protocol. Haematologica 2015; 100:1311-9. [PMID: 26137961 DOI: 10.3324/haematol.2015.126953] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/23/2015] [Indexed: 12/22/2022] Open
Abstract
DNA copy number analysis has been instrumental for the identification of genetic alterations in B-cell precursor acute lymphoblastic leukemia. Notably, some of these genetic defects have been associated with poor treatment outcome and might be relevant for future risk stratification. In this study, we characterized recurrent deletions of CD200 and BTLA genes, mediated by recombination-activating genes, and used breakpoint-specific polymerase chain reaction assay to screen a cohort of 1154 cases of B-cell precursor acute lymphoblastic leukemia uniformly treated according to the EORTC-CLG 58951 protocol. CD200/BTLA deletions were identified in 56 of the patients (4.8%) and were associated with an inferior 8-year event free survival in this treatment protocol [70.2% ± 1.2% for patients with deletions versus 83.5% ± 6.4% for non-deleted cases (hazard ratio 2.02; 95% confidence interval 1.23-3.32; P=0.005)]. Genetically, CD200/BTLA deletions were strongly associated with ETV6-RUNX1-positive leukemias (P<0.0001), but were also identified in patients who did not have any genetic abnormality that is currently used for risk stratification. Within the latter population of patients, the presence of CD200/BTLA deletions was associated with inferior event-free survival and overall survival. Moreover, the multivariate Cox model indicated that these deletions had independent prognostic impact on event-free survival when adjusting for conventional risk criteria. All together, these findings further underscore the rationale for copy number profiling as an important tool for risk stratification in human B-cell precursor acute lymphoblastic leukemia. This trial was registered at www.ClinicalTrials.gov as #NCT00003728.
Collapse
Affiliation(s)
- Farzaneh Ghazavi
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium Center for Medical Genetics, Ghent University Hospital, Belgium
| | - Emmanuelle Clappier
- Department of Genetics, Robert Debré Hospital, APHP, Paris, France Hematology University Institute, University Paris-Diderot, Paris, France
| | - Tim Lammens
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
| | | | - Aurélie Caye
- Department of Genetics, Robert Debré Hospital, APHP, Paris, France Hematology University Institute, University Paris-Diderot, Paris, France
| | - Samira Zegrari
- Department of Genetics, Robert Debré Hospital, APHP, Paris, France
| | - Marleen Bakkus
- Department of Hematology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Belgium
| | - Nathalie Grardel
- Centre de Biologie Pathologie PM Degand, INSERM U837, Lille, France
| | - Yves Benoit
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
| | - Yves Bertrand
- Institute of Hematology and Oncology Paediatrics, Hospices Civils de Lyon, France
| | | | - Vitor Costa
- Department of Pediatrics, Portuguese Oncology Institute, Porto, Portugal
| | - Alina Ferster
- Department of Hemato-Oncology, HUDERF, Brussels, Belgium
| | | | - Geneviève Plat
- Department of Hematology, Children's Hospital, Toulouse, France
| | | | - Marilyne Poirée
- Department of Pediatric Onco-Hematology, Archet University Hospital, Nice, France
| | - Anne Uyttebroeck
- Department of Pediatric Hematology-Oncology, University Hospitals Leuven, Belgium
| | | | - Karima Yakouben
- Department of Pediatric Hematology, Robert Debré Hospital, APHP, Paris, France
| | - Hetty Helsmoortel
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium Center for Medical Genetics, Ghent University Hospital, Belgium
| | - Magali Meul
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
| | - Nadine Van Roy
- Center for Medical Genetics, Ghent University Hospital, Belgium
| | - Jan Philippé
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Belgium
| | - Frank Speleman
- Center for Medical Genetics, Ghent University Hospital, Belgium
| | - Hélène Cavé
- Department of Genetics, Robert Debré Hospital, APHP, Paris, France Hematology University Institute, University Paris-Diderot, Paris, France
| | | | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
| |
Collapse
|