1
|
Li Q, Xing S, Zhang H, Mao X, Xiao M, Wang Y. FISH combined with RT-PCR facilitates classification of Chinese adult patients with B-other ALL through improved identification of ZNF384 rearrangement. Leuk Lymphoma 2024:1-9. [PMID: 39520726 DOI: 10.1080/10428194.2024.2426055] [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: 08/17/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
ZNF384 gene rearrangements are a distinct subtype of adult B cell acute lymphoblastic leukemia (B-ALL). We screened 46 B-other ALL patients for ZNF384 fusions using fluorescent in situ hybridization (FISH) and reverse transcription-polymerase chain reaction (RT-PCR). Clinical data, treatment response, and minimal residual disease (MRD) status were analyzed. Ten (21.7%) patients were ZNF384-r positive (nine by FISH, nine by RT-PCR, eight by both). FISH showed atypical signals, including 3' signal gain and 5' signal deletion. EP300 was the main fusion partner (n = 5). TAF15::ZNF384, SYNRG::ZNF384, CREBBP::ZNF384, and TCF3::ZNF384 fusions were found in one patient each; one case's partner gene is unknown. One patient was MRD-negative at the end of the first induction, lower than in patients without ZNF384-r. ZNF384-r incidence matched B-other ALL incidence in Chinese patients. Combined FISH and RT-PCR improved detection. ALL with ZNF384-r has unique features, and lower MRD-negative response may indicate a negative impact on traditional treatments.
Collapse
Affiliation(s)
- Qinlu Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shugang Xing
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Torres-Diz M, Reglero C, Falkenstein CD, Castro A, Hayer KE, Radens CM, Quesnel-Vallières M, Ang Z, Sehgal P, Li MM, Barash Y, Tasian SK, Ferrando A, Thomas-Tikhonenko A. An Alternatively Spliced Gain-of-Function NT5C2 Isoform Contributes to Chemoresistance in Acute Lymphoblastic Leukemia. Cancer Res 2024; 84:3327-3336. [PMID: 39094066 PMCID: PMC11474164 DOI: 10.1158/0008-5472.can-23-3804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/11/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Relapsed or refractory B-cell acute lymphoblastic leukemia (B-ALL) is a major cause of pediatric cancer-related deaths. Relapse-specific mutations do not account for all chemotherapy failures in B-ALL patients, suggesting additional mechanisms of resistance. By mining RNA sequencing datasets of paired diagnostic/relapse pediatric B-ALL samples, we discovered pervasive alternative splicing (AS) patterns linked to relapse and affecting drivers of resistance to glucocorticoids, antifolates, and thiopurines. Most splicing variations represented cassette exon skipping, "poison" exon inclusion, and intron retention, phenocopying well-documented loss-of-function mutations. In contrast, relapse-associated AS of NT5C2 mRNA yielded an isoform with the functionally uncharacterized in-frame exon 6a. Incorporation of the 8-amino acid sequence SQVAVQKR into this enzyme created a putative phosphorylation site and resulted in elevated nucleosidase activity, which is a known consequence of gain-of-function mutations in NT5C2 and a common determinant of 6-mercaptopurine resistance. Consistent with this finding, NT5C2ex6a and the R238W hotspot variant conferred comparable levels of resistance to 6-mercaptopurine in B-ALL cells both in vitro and in vivo. Furthermore, both NT5C2ex6a and the R238W variant induced collateral sensitivity to the inosine monophosphate dehydrogenase inhibitor mizoribine. These results ascribe to splicing perturbations an important role in chemotherapy resistance in relapsed B-ALL and suggest that inosine monophosphate dehydrogenase inhibitors, including the commonly used immunosuppressive agent mycophenolate mofetil, could be a valuable therapeutic option for treating thiopurine-resistant leukemias. Significance: Alternative splicing is a potent mechanism of acquired drug resistance in relapsed/refractory acute lymphoblastic leukemias that has diagnostic and therapeutic implications for patients who lack mutations in known chemoresistance genes.
Collapse
Affiliation(s)
- Manuel Torres-Diz
- Division of Cancer Pathobiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Clara Reglero
- Institute for Cancer Genetics, Columbia University, New York, New York.
| | | | - Annette Castro
- Division of Cancer Pathobiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Katharina E. Hayer
- Division of Cancer Pathobiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Caleb M. Radens
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Mathieu Quesnel-Vallières
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Zhiwei Ang
- Division of Cancer Pathobiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Priyanka Sehgal
- Division of Cancer Pathobiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Marilyn M. Li
- Division of Genomic Diagnostic, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Yoseph Barash
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Sarah K. Tasian
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University, New York, New York.
- Department of Pediatrics, Columbia University, New York, New York.
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania.
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
3
|
Chang TC, Chen W, Qu C, Cheng Z, Hedges D, Elsayed A, Pounds SB, Shago M, Rabin KR, Raetz EA, Devidas M, Cheng C, Angiolillo A, Baviskar P, Borowitz M, Burke MJ, Carroll A, Carroll WL, Chen IM, Harvey R, Heerema N, Iacobucci I, Wang JR, Jeha S, Larsen E, Mattano L, Maloney K, Pui CH, Ramirez NC, Salzer W, Willman C, Winick N, Wood B, Hunger SP, Wu G, Mullighan CG, Loh ML. Genomic Determinants of Outcome in Acute Lymphoblastic Leukemia. J Clin Oncol 2024; 42:3491-3503. [PMID: 39121442 PMCID: PMC11458106 DOI: 10.1200/jco.23.02238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/23/2024] [Accepted: 05/31/2024] [Indexed: 08/11/2024] Open
Abstract
PURPOSE Although cure rates for childhood acute lymphoblastic leukemia (ALL) exceed 90%, ALL remains a leading cause of cancer death in children. Half of relapses arise in children initially classified with standard-risk (SR) disease. MATERIALS AND METHODS To identify genomic determinants of relapse in children with SR ALL, we performed genome and transcriptome sequencing of diagnostic and remission samples of children with SR (n = 1,381) or high-risk B-ALL with favorable cytogenetic features (n = 115) enrolled on Children's Oncology Group trials. We used a case-control study design analyzing 439 patients who relapsed and 1,057 who remained in complete remission for at least 5 years. RESULTS Genomic subtype was associated with relapse, which occurred in approximately 50% of cases of PAX5-altered ALL (odds ratio [OR], 3.31 [95% CI, 2.17 to 5.03]; P = 3.18 × 10-8). Within high-hyperdiploid ALL, gain of chromosome 10 with disomy of chromosome 7 was associated with favorable outcome (OR, 0.27 [95% CI, 0.17 to 0.42]; P = 8.02 × 10-10; St Jude Children's Research Hospital validation cohort: OR, 0.22 [95% CI, 0.05 to 0.80]; P = .009), and disomy of chromosomes 10 and 17 with gain of chromosome 6 was associated with relapse (OR, 7.16 [95% CI, 2.63 to 21.51]; P = 2.19 × 10-5; validation cohort: OR, 21.32 [95% CI, 3.62 to 119.30]; P = .0004). Genomic alterations were associated with relapse in a subtype-dependent manner, including alterations of INO80 in ETV6::RUNX1 ALL, IKZF1, and CREBBP in high-hyperdiploid ALL and FHIT in BCR::ABL1-like ALL. Genomic alterations were also associated with the presence of minimal residual disease, including NRAS and CREBBP in high-hyperdiploid ALL. CONCLUSION Genetic subtype, patterns of aneuploidy, and secondary genomic alterations determine risk of relapse in childhood ALL. Comprehensive genomic analysis is required for optimal risk stratification.
Collapse
Affiliation(s)
- Ti-Cheng Chang
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, TN
| | - Wenan Chen
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, TN
- Division of Computational Biology, Mayo Clinic, Rochester, MN
| | - Chunxu Qu
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN
| | - Zhongshan Cheng
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, TN
| | - Dale Hedges
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, TN
- Deceased
| | - Abdelrahman Elsayed
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN
| | - Stanley B. Pounds
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN
| | - Mary Shago
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Karen R. Rabin
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Elizabeth A. Raetz
- Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Hospital, New York, NY
| | - Meenakshi Devidas
- Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| | - Cheng Cheng
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN
| | | | | | - Michael Borowitz
- Department of Pathology, Johns Hopkins University, Baltimore, MD
| | - Michael J. Burke
- Division of Pediatric Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Andrew Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL
| | - William L. Carroll
- Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Hospital, New York, NY
| | - I-Ming Chen
- Department of Pathology, University of New Mexico, Albuquerque, NM
| | - Richard Harvey
- Department of Pathology, University of New Mexico, Albuquerque, NM
| | | | - Ilaria Iacobucci
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN
| | - Jeremy R. Wang
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sima Jeha
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Eric Larsen
- Department of Pediatrics, Maine Children's Cancer Program, Scarborough, ME
| | | | - Kelly Maloney
- Department of Pediatrics and Children's Hospital Colorado, University of Colorado, Aurora, CO
| | - Ching-Hon Pui
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Nilsa C. Ramirez
- Departments of Pathology and Pediatrics, Institute for Genomic Medicine and Biopathology Center, Nationwide Children's Hospital, Ohio State University, Columbus, OH
| | - Wanda Salzer
- Uniformed Services University, School of Medicine, Bethesda, MD
| | - Cheryl Willman
- Department of Laboratory Medicine and Pathology and Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, MN
| | - Naomi Winick
- Department of Pediatric Hematology Oncology and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Brent Wood
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA
| | - Stephen P. Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Gang Wu
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, TN
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN
| | | | - Mignon L. Loh
- Department of Pediatrics and the Ben Towne Center for Childhood Cancer Research, Seattle Children's Hospital, University of Washington, Seattle, WA
| |
Collapse
|
4
|
Li L, Xiao H, Wu X, Tang Z, Khoury JD, Wang J, Wan S. RanBALL: An Ensemble Random Projection Model for Identifying Subtypes of B-cell Acute Lymphoblastic Leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614777. [PMID: 39386448 PMCID: PMC11463541 DOI: 10.1101/2024.09.24.614777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
As the most common pediatric malignancy, B-cell acute lymphoblastic leukemia (B-ALL) has multiple distinct subtypes characterized by recurrent and sporadic somatic and germline genetic alterations. Identification of B-ALL subtypes can facilitate risk stratification and enable tailored therapeutic approaches. Existing methods for B-ALL subtyping primarily depend on immunophenotypic, cytogenetic and genomic analyses, which would be costly, complicated, and laborious in clinical practice applications. To overcome these challenges, we present RanBALL (an Ensemble Random Projection-Based Model for Identifying B-Cell Acute Lymphoblastic Leukemia Subtypes), an accurate and cost-effective model for B-ALL subtype identification based on transcriptomic profiling only. RanBALL leverages random projection (RP) to construct an ensemble of dimension-reduced multi-class support vector machine (SVM) classifiers for B-ALL subtyping. Results based on 100 times 5-fold cross validation tests for >1700 B-ALL patients demonstrated that the proposed model achieved an accuracy of 93.35%, indicating promising prediction capabilities of RanBALL for B-ALL subtyping. The high accuracies of RanBALL suggested that our model could effectively capture underlying patterns of transcriptomic profiling for accurate B-ALL subtype identification. We believe RanBALL will facilitate the discovery of B-ALL subtype-specific marker genes and therapeutic targets, and eventually have consequential positive impacts on downstream risk stratification and tailored treatment design.
Collapse
Affiliation(s)
- Lusheng Li
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hanyu Xiao
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xinchao Wu
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Zhenya Tang
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joseph D. Khoury
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jieqiong Wang
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shibiao Wan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
5
|
Kolekar P, Balagopal V, Dong L, Liu Y, Foy S, Tran Q, Mulder H, Huskey ALW, Plyler E, Liang Z, Ma J, Nakitandwe J, Gu J, Namwanje M, Maciaszek J, Payne-Turner D, Mallampati S, Wang L, Easton J, Klco JM, Ma X. SJPedPanel: A Pan-Cancer Gene Panel for Childhood Malignancies to Enhance Cancer Monitoring and Early Detection. Clin Cancer Res 2024; 30:4100-4114. [PMID: 39047169 PMCID: PMC11393547 DOI: 10.1158/1078-0432.ccr-24-1063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/14/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE The purpose of the study was to design a pan-cancer gene panel for childhood malignancies and validate it using clinically characterized patient samples. EXPERIMENTAL DESIGN In addition to 5,275 coding exons, SJPedPanel also covers 297 introns for fusions/structural variations and 7,590 polymorphic sites for copy-number alterations. Capture uniformity and limit of detection are determined by targeted sequencing of cell lines using dilution experiment. We validate its coverage by in silico analysis of an established real-time clinical genomics (RTCG) cohort of 253 patients. We further validate its performance by targeted resequencing of 113 patient samples from the RTCG cohort. We demonstrate its power in analyzing low tumor burden specimens using morphologic remission and monitoring samples. RESULTS Among the 485 pathogenic variants reported in RTCG cohort, SJPedPanel covered 86% of variants, including 82% of 90 rearrangements responsible for fusion oncoproteins. In our targeted resequencing cohort, 91% of 389 pathogenic variants are detected. The gene panel enabled us to detect ∼95% of variants at allele fraction (AF) 0.5%, whereas the detection rate is ∼80% at AF 0.2%. The panel detected low-frequency driver alterations from morphologic leukemia remission samples and relapse-enriched alterations from monitoring samples, demonstrating its power for cancer monitoring and early detection. CONCLUSIONS SJPedPanel enables the cost-effective detection of clinically relevant genetic alterations including rearrangements responsible for subtype-defining fusions by targeted sequencing of ∼0.15% of human genome for childhood malignancies. It will enhance the analysis of specimens with low tumor burdens for cancer monitoring and early detection.
Collapse
Affiliation(s)
- Pandurang Kolekar
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Vidya Balagopal
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Li Dong
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yanling Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Scott Foy
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Quang Tran
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Heather Mulder
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Anna L W Huskey
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Emily Plyler
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Zhikai Liang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jingqun Ma
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Joy Nakitandwe
- Department of Pathology and Laboratory Medicine, Diagnostics Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jiali Gu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Maria Namwanje
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jamie Maciaszek
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Debbie Payne-Turner
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Saradhi Mallampati
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Lu Wang
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
6
|
Østergaard A, Boer JM, van Leeuwen FN, Pieters R, Den Boer ML. IKZF1 in acute lymphoblastic leukemia: the rise before the fall? Leuk Lymphoma 2024:1-11. [PMID: 39210599 DOI: 10.1080/10428194.2024.2396046] [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: 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
|
7
|
Bernt KM. Rethinking paraneoplastic eosinophilia. Haematologica 2024; 109:2385-2387. [PMID: 38572550 PMCID: PMC11290496 DOI: 10.3324/haematol.2024.285081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Kathrin M Bernt
- Division of Pediatric Oncology, Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Abramson Cancer Center, Philadelphia, PA.
| |
Collapse
|
8
|
Rujkijyanont P, Inaba H. Diagnostic and treatment strategies for pediatric acute lymphoblastic leukemia in low- and middle-income countries. Leukemia 2024; 38:1649-1662. [PMID: 38762553 DOI: 10.1038/s41375-024-02277-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/20/2024]
Abstract
The survival rate of children and adolescents with acute lymphoblastic leukemia (ALL), the most common pediatric cancer, has improved significantly in high-income countries (HICs), serving as an excellent example of how humans can overcome catastrophic diseases. However, the outcomes in children with ALL in low- and middle-income countries (LMICs), where approximately 80% of the global population live, are suboptimal because of limited access to diagnostic procedures, chemotherapeutic agents, supportive care, and financial assistance. Although the implementation of therapeutic strategies in resource-limited countries could theoretically follow the same path of improvement as modeled in HICs, intensification of chemotherapy may simply result in increased toxicities. With the advent of genetic diagnosis, molecular targeted therapy, and immunotherapy, the management of ALL is changing dramatically in HICs. Multidisciplinary collaborations between institutions in LMICs and HICs will provide access to strategies that are suitable for institutions in LMICs, enabling them to minimize toxicities while improving outcomes. This article summarizes important aspects of the diagnosis and treatment of pediatric ALL that were mostly developed in HICs but that can be realistically implemented by institutions in countries with limited resources through resource-adapted multidisciplinary collaborations.
Collapse
Affiliation(s)
- Piya Rujkijyanont
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao Hospital and Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Hiroto Inaba
- Leukemia/Lymphoma Division, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
9
|
Litzow MR, Sun Z, Mattison RJ, Paietta EM, Roberts KG, Zhang Y, Racevskis J, Lazarus HM, Rowe JM, Arber DA, Wieduwilt MJ, Liedtke M, Bergeron J, Wood BL, Zhao Y, Wu G, Chang TC, Zhang W, Pratz KW, Dinner SN, Frey N, Gore SD, Bhatnagar B, Atallah EL, Uy GL, Jeyakumar D, Lin TL, Willman CL, DeAngelo DJ, Patel SB, Elliott MA, Advani AS, Tzachanis D, Vachhani P, Bhave RR, Sharon E, Little RF, Erba HP, Stone RM, Luger SM, Mullighan CG, Tallman MS. Blinatumomab for MRD-Negative Acute Lymphoblastic Leukemia in Adults. N Engl J Med 2024; 391:320-333. [PMID: 39047240 PMCID: PMC11334054 DOI: 10.1056/nejmoa2312948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
BACKGROUND Many older adults with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) have a relapse despite having a measurable residual disease (MRD)-negative complete remission with combination chemotherapy. The addition of blinatumomab, a bispecific T-cell engager molecule that is approved for the treatment of relapsed, refractory, and MRD-positive BCP-ALL, may have efficacy in patients with MRD-negative remission. METHODS In a phase 3 trial, we randomly assigned patients 30 to 70 years of age with BCR::ABL1-negative BCP-ALL (with :: indicating fusion) who had MRD-negative remission (defined as <0.01% leukemic cells in bone marrow as assessed on flow cytometry) after induction and intensification chemotherapy to receive four cycles of blinatumomab in addition to four cycles of consolidation chemotherapy or to receive four cycles of consolidation chemotherapy alone. The primary end point was overall survival, and relapse-free survival was a secondary end point. RESULTS The data and safety monitoring committee reviewed the results from the third efficacy interim analysis and recommended that they be reported. Complete remission with or without full count recovery was observed in 395 of 488 enrolled patients (81%). Of the 224 patients with MRD-negative status, 112 were assigned to each group. The characteristics of the patients were balanced between the groups. At a median follow-up of 43 months, an advantage was observed in the blinatumomab group as compared with the chemotherapy-only group with regard to overall survival (at 3 years: 85% vs. 68%; hazard ratio for death, 0.41; 95% confidence interval [CI], 0.23 to 0.73; P = 0.002), and the 3-year relapse-free survival was 80% with blinatumomab and 64% with chemotherapy alone (hazard ratio for relapse or death, 0.53; 95% CI, 0.32 to 0.87). A higher incidence of neuropsychiatric events was reported in the blinatumomab group than in the chemotherapy-only group. CONCLUSIONS The addition of blinatumomab to consolidation chemotherapy in adult patients in MRD-negative remission from BCP-ALL significantly improved overall survival. (Funded by the National Institutes of Health and others; E1910 ClinicalTrials.gov number, NCT02003222.).
Collapse
Affiliation(s)
- Mark R Litzow
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Zhuoxin Sun
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Ryan J Mattison
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Elisabeth M Paietta
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Kathryn G Roberts
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Yanming Zhang
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Janis Racevskis
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Hillard M Lazarus
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Jacob M Rowe
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Daniel A Arber
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Matthew J Wieduwilt
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Michaela Liedtke
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Julie Bergeron
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Brent L Wood
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Yaqi Zhao
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Gang Wu
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Ti-Cheng Chang
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Wenchao Zhang
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Keith W Pratz
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Shira N Dinner
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Noelle Frey
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Steven D Gore
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Bhavana Bhatnagar
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Ehab L Atallah
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Geoffrey L Uy
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Deepa Jeyakumar
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Tara L Lin
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Cheryl L Willman
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Daniel J DeAngelo
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Shejal B Patel
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Michelle A Elliott
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Anjali S Advani
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Dimitrios Tzachanis
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Pankit Vachhani
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Rupali R Bhave
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Elad Sharon
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Richard F Little
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Harry P Erba
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Richard M Stone
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Selina M Luger
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Charles G Mullighan
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| | - Martin S Tallman
- From the Mayo Clinic, Rochester, MN (M.R.L., C.L.W., M.A.E.); Dana-Farber Cancer Institute, Boston (Z.S., D.J.D., R.M.S.); the University of Wisconsin Carbone Cancer Center, Madison (R.J.M.), and the Medical College of Wisconsin, Milwaukee (E.L.A.); Montefiore Medical Center Moses Campus (E.M.P., J.R.) and Memorial Sloan Kettering Cancer Center (Y. Zhang, M.S.T.) - both in New York; the Department of Pathology and the Center for Excellence for Leukemia Studies (K.G.R., Y. Zhao, C.G.M.) and the Center for Applied Bioinformatics (G.W., T.-C.C., W.Z.), St. Jude's Children's Research Hospital, Memphis, TN; Case Western Reserve University (H.M.L.) and Cleveland Clinic Foundation (A.S.A.), Cleveland, and the Ohio State University Comprehensive Cancer Center, Columbus (B.B.) - all in Ohio; Shaare Zedek Medical Center, Jerusalem, Israel (J.M.R.); Stanford Cancer Institute, Palo Alto (D.A.A., M.L.), the University of California, San Diego, Moores Cancer Center, La Jolla (M.J.W., D.T.), and the University of California, Irvine, Health Cancer Center-Newport, Orange (D.J.) - all in California; the University of Chicago (D.A.A.) and Northwestern University (S.N.D.) - both in Chicago; Hopital Maisonneuve-Rosemont, Montreal (J.B.); the University of Washington, Seattle (B.L.W.); Johns Hopkins University Sidney Kimmel Cancer Center, Baltimore (K.W.P.), and the National Cancer Institute, National Institutes of Health, Bethesda (E.S., R.F.L.) - both in Maryland; the University of Pennsylvania Abramson Cancer Center, Philadelphia (N.F., S.M.L.); Yale School of Medicine, New Haven, CT (S.D.G.); the Washington University in St. Louis School of Medicine, St. Louis (G.L.U.); the University of Kansas Cancer Center, Westwood (T.L.L.); Virginia Commonwealth University Massey Cancer Center, Richmond (S.B.P.); the University of Alabama at Birmingham, Birmingham (P.V.); and Wake Forest University Health Sciences, Winston-Salem (R.R.B.), and Duke University Medical Center, Durham (H.P.E.) - both in North Carolina
| |
Collapse
|
10
|
Torres-Diz M, Reglero C, Falkenstein CD, Castro A, Hayer KE, Radens CM, Quesnel-Vallières M, Ang Z, Sehgal P, Li MM, Barash Y, Tasian SK, Ferrando A, Thomas-Tikhonenko A. An Alternatively Spliced Gain-of-Function NT5C2 Isoform Contributes to Chemoresistance in Acute Lymphoblastic Leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.14.557413. [PMID: 39091882 PMCID: PMC11291008 DOI: 10.1101/2023.09.14.557413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Relapsed or refractory B-cell acute lymphoblastic leukemia (B-ALL) is a major cause of pediatric cancer-related deaths. Relapse-specific mutations do not account for all chemotherapy failures in B- ALL patients, suggesting additional mechanisms of resistance. By mining RNA-seq datasets of paired diagnostic/relapse pediatric B-ALL samples, we discovered pervasive alternative splicing (AS) patterns linked to relapse and affecting drivers of resistance to glucocorticoids, anti-folates, and thiopurines. Most splicing variations represented cassette exon skipping, "poison" exon inclusion, and intron retention, phenocopying well-documented loss-of-function mutations. In contrast, relapse-associated AS of NT5C2 mRNA yielded an isoform with the functionally uncharacterized in-frame exon 6a. Incorporation of the 8-amino acid sequence SQVAVQKR into this enzyme created a putative phosphorylation site and resulted in elevated nucleosidase activity, which is a known consequence of gain-of-function mutations in NT5C2 and a common determinant of 6-mercaptopurine (6-MP) resistance. Consistent with this finding, NT5C2ex6a and the R238W hotspot variant conferred comparable levels of resistance to 6-MP in B-ALL cells both in vitro and in vivo. Furthermore, both the NT5C2ex6a and R238W variants induced collateral sensitivity to the inosine monophosphate dehydrogenase (IMPDH) inhibitor mizoribine. These results ascribe an important role for splicing perturbations in chemotherapy resistance in relapsed B-ALL and suggest that IMPDH inhibitors, including the commonly used immunosuppressive agent mycophenolate mofetil, could be a valuable therapeutic option for treating thiopurine-resistant leukemias.
Collapse
|
11
|
Schinnerl D, Riebler M, Schumich A, Haslinger S, Bramböck A, Inthal A, Nykiel M, Maurer-Granofszky M, Haas OA, Pötschger U, Köhrer S, Nebral K, Dworzak MN, Attarbaschi A, Strehl S. Risk factors in DUX4-positive childhood and adolescent B-cell acute lymphoblastic leukemia. Blood Cancer J 2024; 14:119. [PMID: 39039054 PMCID: PMC11263571 DOI: 10.1038/s41408-024-01099-3] [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: 05/07/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Affiliation(s)
- Dagmar Schinnerl
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Marion Riebler
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Angela Schumich
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Sabrina Haslinger
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Alice Bramböck
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Andrea Inthal
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Marek Nykiel
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Margarita Maurer-Granofszky
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Oskar A Haas
- St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Ulrike Pötschger
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Stefan Köhrer
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Karin Nebral
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Michael N Dworzak
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
- St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Andishe Attarbaschi
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Sabine Strehl
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
| |
Collapse
|
12
|
Hodder A, Leiter SM, Kennedy J, Addy D, Ahmed M, Ajithkumar T, Allinson K, Ancliff P, Bailey S, Barnard G, Burke GAA, Burns C, Cano-Flanagan J, Chalker J, Coleman N, Cheng D, Clinch Y, Dryden C, Ghorashian S, Griffin B, Horan G, Hubank M, May P, McDerra J, Nagrecha R, Nicholson J, O'Connor D, Pavasovic V, Quaegebeur A, Rao A, Roberts T, Samarasinghe S, Stasevich I, Tadross JA, Trayers C, Trotman J, Vora A, Watkins J, Chitty LS, Bowdin S, Armstrong R, Murray MJ, Hook CE, Tarpey P, Vedi A, Bartram J, Behjati S. Benefits for children with suspected cancer from routine whole-genome sequencing. Nat Med 2024; 30:1905-1912. [PMID: 38956197 PMCID: PMC11271414 DOI: 10.1038/s41591-024-03056-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/08/2024] [Indexed: 07/04/2024]
Abstract
Clinical whole-genome sequencing (WGS) has been shown to deliver potential benefits to children with cancer and to alter treatment in high-risk patient groups. It remains unknown whether offering WGS to every child with suspected cancer can change patient management. We collected WGS variant calls and clinical and diagnostic information from 281 children (282 tumors) across two English units (n = 152 from a hematology center, n = 130 from a solid tumor center) where WGS had become a routine test. Our key finding was that variants uniquely attributable to WGS changed the management in ~7% (20 out of 282) of cases while providing additional disease-relevant findings, beyond standard-of-care molecular tests, in 108 instances for 83 (29%) cases. Furthermore, WGS faithfully reproduced every standard-of-care molecular test (n = 738) and revealed several previously unknown genomic features of childhood tumors. We show that WGS can be delivered as part of routine clinical care to children with suspected cancer and can change clinical management by delivering unexpected genomic insights. Our experience portrays WGS as a clinically impactful assay for routine practice, providing opportunities for assay consolidation and for delivery of molecularly informed patient care.
Collapse
Affiliation(s)
- Angus Hodder
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Sarah M Leiter
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Jonathan Kennedy
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Dilys Addy
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Munaza Ahmed
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Kieren Allinson
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Phil Ancliff
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Shivani Bailey
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Gemma Barnard
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - G A Amos Burke
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Charlotte Burns
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | - Nicholas Coleman
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Danny Cheng
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Caryl Dryden
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Sara Ghorashian
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Blanche Griffin
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- North Thames Genomic Laboratory Hub, London, UK
| | - Gail Horan
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Michael Hubank
- North Thames Genomic Laboratory Hub, London, UK
- The Institute of Cancer Research, London, UK
| | - Phillippa May
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Joanna McDerra
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Rajvi Nagrecha
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - James Nicholson
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - David O'Connor
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - Vesna Pavasovic
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Annelies Quaegebeur
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Anupama Rao
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Thomas Roberts
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
| | | | - Iryna Stasevich
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - John A Tadross
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Claire Trayers
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Jamie Trotman
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
| | - Ajay Vora
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - James Watkins
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
| | - Lyn S Chitty
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- North Thames Genomic Laboratory Hub, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sarah Bowdin
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
| | - Ruth Armstrong
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Matthew J Murray
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Catherine E Hook
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Patrick Tarpey
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- East Genomics Laboratory Hub, Cambridge, UK.
| | - Aditi Vedi
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| | - Jack Bartram
- Great Ormond Street Hospital NHS Foundation Trust, London, UK.
- North Thames Genomic Laboratory Hub, London, UK.
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Hu Z, Jia Z, Liu J, Mao A, Han H, Gu Z. MD-ALL: an integrative platform for molecular diagnosis of B-acute lymphoblastic leukemia. Haematologica 2024; 109:1741-1754. [PMID: 37981856 PMCID: PMC11141650 DOI: 10.3324/haematol.2023.283706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023] Open
Abstract
B-acute lymphoblastic leukemia (B-ALL) consists of dozens of subtypes defined by distinct gene expression profiles (GEP) and various genetic lesions. With the application of transcriptome sequencing (RNA sequencing [RNA-seq]), multiple novel subtypes have been identified, which lead to an advanced B-ALL classification and risk-stratification system. However, the complexity of analyzing RNA-seq data for B-ALL classification hinders the implementation of the new B-ALL taxonomy. Here, we introduce Molecular Diagnosis of Acute Lymphoblastic Leukemia (MD-ALL), an integrative platform featuring sensitive and accurate B-ALL classification based on GEP and sentinel genetic alterations from RNA-seq data. In this study, we systematically analyzed 2,955 B-ALL RNA-seq samples and generated a reference dataset representing all the reported B-ALL subtypes. Using multiple machine learning algorithms, we identified the feature genes and then established highly sensitive and accurate models for B-ALL classification using either bulk or single-cell RNA-seq data. Importantly, this platform integrates multiple aspects of key genetic lesions acquired from RNA-seq data, which include sequence mutations, large-scale copy number variations, and gene rearrangements, to perform comprehensive and definitive B-ALL classification. Through validation in a hold-out cohort of 974 samples, our models demonstrated superior performance for B-ALL classification compared with alternative tools. Moreover, to ensure accessibility and user-friendly navigation even for users with limited or no programming background, we developed an interactive graphical user interface for this MD-ALL platform, using the R Shiny package. In summary, MD-ALL is a user-friendly B-ALL classification platform designed to enable integrative, accurate, and comprehensive B-ALL subtype classification. MD-ALL is available from https://github.com/gu-lab20/MD-ALL.
Collapse
Affiliation(s)
- Zunsong Hu
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA
| | - Zhilian Jia
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA
| | - Jiangyue Liu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA; Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA
| | - Allen Mao
- Research Informatics, City of Hope National Medical Center, Duarte, CA
| | - Helen Han
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA
| | - Zhaohui Gu
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA.
| |
Collapse
|
14
|
Tamefusa K, Ochi M, Ishida H, Shiwaku T, Kanamitsu K, Fujiwara K, Tatebe Y, Matsumoto N, Washio K, Tsukahara H. Delayed diagnostic interval and survival outcomes in pediatric leukemia: A single-center, retrospective study. Eur J Haematol 2024; 112:714-722. [PMID: 38152024 DOI: 10.1111/ejh.14162] [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: 08/03/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE This study primarily focused on the diagnostic interval (DI), defined as the duration from the onset of leukemic symptoms to diagnosis. We investigated whether a prolonged DI is associated with the outcomes of pediatric leukemia. METHODS We retrospectively collected data of children with newly diagnosed pediatric leukemia at Okayama University Hospital from January 2007 to December 2022. Survival analyses were conducted using Kaplan-Meier methods, and an unadjusted analysis to compare differences in survival was performed using the log-rank test. RESULTS In total, 103 children with leukemia were included in the analysis. The median DI was 20 days (interquartile range, 9.5-33.5 days). A prolonged DI (≥30 days) demonstrated no association with either 5-year event-free survival (70.1% for <30 days and 68.3% for ≥30 days, p = .99, log-rank test) or overall survival (84.7% for <30 days and 89.4% for ≥30 days, p = .85, log-rank test). CONCLUSIONS A prolonged DI was not associated with the survival of children with leukemia. If a precise classification of leukemia biology is provided for pediatric patients, a prolonged DI may have little impact on the prognosis of these patients.
Collapse
Affiliation(s)
- Kosuke Tamefusa
- Department of Pediatrics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Motoharu Ochi
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Hisashi Ishida
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Takahiro Shiwaku
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Kiichiro Kanamitsu
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
- Department of Pediatrics, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Kaori Fujiwara
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Yasuhisa Tatebe
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Naomi Matsumoto
- Department of Epidemiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kana Washio
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Hirokazu Tsukahara
- Department of Pediatrics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
15
|
Choi JK, Xiao W, Chen X, Loghavi S, Elenitoba-Johnson KS, Naresh KN, Medeiros LJ, Czader M. Fifth Edition of the World Health Organization Classification of Tumors of the Hematopoietic and Lymphoid Tissues: Acute Lymphoblastic Leukemias, Mixed-Phenotype Acute Leukemias, Myeloid/Lymphoid Neoplasms With Eosinophilia, Dendritic/Histiocytic Neoplasms, and Genetic Tumor Syndromes. Mod Pathol 2024; 37:100466. [PMID: 38460674 DOI: 10.1016/j.modpat.2024.100466] [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: 10/06/2023] [Revised: 02/11/2024] [Accepted: 03/01/2024] [Indexed: 03/11/2024]
Abstract
This manuscript represents a review of lymphoblastic leukemia/lymphoma (acute lymphoblastic leukemia/lymphoblastic lymphoma), acute leukemias of ambiguous lineage, mixed-phenotype acute leukemias, myeloid/lymphoid neoplasms with eosinophilia and defining gene rearrangements, histiocytic and dendritic neoplasms, and genetic tumor syndromes of the 5th edition of the World Health Organization Classification of Tumors of the Hematopoietic and Lymphoid Tissues. The diagnostic, clinicopathologic, cytogenetic, and molecular genetic features are discussed. The differences in comparison to the 4th revised edition of the World Health Organization classification of hematolymphoid neoplasms are highlighted.
Collapse
Affiliation(s)
- John K Choi
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wenbin Xiao
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xueyan Chen
- Section of Pathology, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Sanam Loghavi
- Department of Hematopathology, MD Anderson Cancer Center, Houston, Texas
| | - Kojo S Elenitoba-Johnson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kikkeri N Naresh
- Section of Pathology, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - L Jeffrey Medeiros
- Department of Hematopathology, MD Anderson Cancer Center, Houston, Texas
| | - Magdalena Czader
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
16
|
Buldini B, Varotto E, Maurer-Granofszky M, Gaipa G, Schumich A, Brüggemann M, Mejstrikova E, Cazzaniga G, Hrusak O, Szczepanowski M, Scarparo P, Zimmermann M, Strehl S, Schinnerl D, Zaliova M, Karawajew L, Bourquin JP, Feuerstein T, Cario G, Alten J, Möricke A, Biffi A, Parasole R, Fagioli F, Valsecchi MG, Biondi A, Locatelli F, Attarbaschi A, Schrappe M, Conter V, Basso G, Dworzak MN. CD371-positive pediatric B-cell acute lymphoblastic leukemia: propensity to lineage switch and slow early response to treatment. Blood 2024; 143:1738-1751. [PMID: 38215390 DOI: 10.1182/blood.2023021952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/27/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024] Open
Abstract
ABSTRACT In the effort to improve immunophenotyping and minimal residual disease (MRD) assessment in acute lymphoblastic leukemia (ALL), the international Berlin-Frankfurt-Münster (iBFM) Flow Network introduced the myelomonocytic marker CD371 for a large prospective characterization with a long follow-up. In the present study, we aimed to investigate the clinical and biological features of CD371-positive (CD371pos) pediatric B-cell precursor ALL (BCP-ALL). From June 2014 to February 2017, 1812 pediatric patients with newly diagnosed BCP-ALLs enrolled in trial AIEOP-BFM ALL 2009 were evaluated as part of either a screening (n = 843, Italian centers) or validation cohort (n = 969, other iBFM centers). Laboratory assessment at diagnosis consisted of morphological, immunophenotypic, and genetic analysis. Response assessment relied on morphology, multiparametric flow cytometry (MFC), and polymerase chain reaction (PCR)-MRD. At diagnosis, 160 of 1812 (8.8%) BCP-ALLs were CD371pos. This correlated with older age, lower ETV6::RUNX1 frequency, immunophenotypic immaturity (all P < .001), and strong expression of CD34 and of CD45 (P < .05). During induction therapy, CD371pos BCP-ALLs showed a transient myelomonocytic switch (mm-SW: up to 65.4% of samples at day 15) and an inferior response to chemotherapy (slow early response, P < .001). However, the 5-year event-free survival was 88.3%. Among 420 patients from the validation cohort, 27 of 28 (96.4%) cases positive for DUX4-fusions were CD371pos. In conclusion, in the largest pediatric cohort, CD371 is the most sensitive marker of transient mm-SW, whose recognition is essential for proper MFC MRD assessment. CD371pos is associated to poor early treatment response, although a good outcome can be reached after MRD-based ALL-related therapies.
Collapse
Affiliation(s)
- Barbara Buldini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padua, Padua, Italy
- Pediatric Onco-Hematology, Stem Cell Transplant and Gene Therapy Laboratory, Istituto di Ricerca Pediatrica, Città della Speranza, Padua, Italy
| | - Elena Varotto
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padua, Padua, Italy
| | | | - Giuseppe Gaipa
- Tettamanti Center, IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Angela Schumich
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Monika Brüggemann
- Department of Internal Medicine I, Hematology Laboratory, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Ester Mejstrikova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Giovanni Cazzaniga
- Tettamanti Center, IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Ondrej Hrusak
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Monika Szczepanowski
- Department of Internal Medicine I, Hematology Laboratory, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Pamela Scarparo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padua, Padua, Italy
| | | | - Sabine Strehl
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | | | - Marketa Zaliova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Leonid Karawajew
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin, Berlin, Germany
| | - Jean-Pierre Bourquin
- Division of Oncology and Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Tamar Feuerstein
- Immune Phenotype Laboratory, Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Gunnar Cario
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Julia Alten
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Anja Möricke
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padua, Padua, Italy
- Pediatric Onco-Hematology, Stem Cell Transplant and Gene Therapy Laboratory, Istituto di Ricerca Pediatrica, Città della Speranza, Padua, Italy
| | - Rosanna Parasole
- Department of Oncology, Hematology and Cellular Therapy, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Franca Fagioli
- Pediatric Onco-Hematology, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | | | - Andrea Biondi
- Pediatrics, IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology, Cell and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital, Rome, Italy
- Department of Health Science and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Martin Schrappe
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Kiel, Germany
| | | | - Giuseppe Basso
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padua, Padua, Italy
| | - Michael N Dworzak
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Yu CH, Su YH, Jou ST, Lu MY, Lin CY, Lin KH, Chang HH, Chou SW, Huang YH, Lin DT, Lin SW, Chen HY, Chang YH, Yang YL. Terminal deoxynucleotidyl transferase expression in different subtypes of childhood B-cell acute lymphoblastic leukemia. Pathol Res Pract 2024; 256:155287. [PMID: 38579576 DOI: 10.1016/j.prp.2024.155287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/21/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
The lack of expression of terminal deoxynucleotidyl transferase (TdT) is frequently associated with KMT2A-rearranged subtype of pediatric acute lymphoblastic leukemia (ALL). However, this association has not been investigated extensively in the Asian population. A retrospective analysis of TdT expression in pediatric B-cell ALL (B-ALL) was performed in patients treated using the Taiwan Pediatric Oncology Group (TPOG) ALL 2002 and 2013 protocols. Among the 331 patients with B-ALL, 12 patients showed TdT negativity at initial diagnosis. Among these, eight patients showed KMT2A rearrangement (66.7%). Other patients showing negative TdT expression had ETV6::RUNX1, MEF2D-rearranged, and other B-ALL subtypes. However, in the context of KMT2A-rearranged B-ALL (n = 20), only eight patients showed TdT negativity. The 5-year event-free survival and overall survival of patients with and without TdT expression were 83.8% versus 46.8% (P <0.001) and 86.3% versus 55.4% (P = 0.004), respectively. Moreover, several aberrant markers, such as CD2, CD56, CD7, and CD117, were rarely expressed in the B-ALL samples, and if expressed, they were enriched in specific genetic subtypes. The results of this study indicate that immunophenotypic features are correlated with specific genetic subtypes of childhood B-ALL.
Collapse
Affiliation(s)
- Chih-Hsiang Yu
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan; Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | | | - Shiann-Tarng Jou
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Yao Lu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Yu Lin
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Kai-Hsin Lin
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiu-Hao Chang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Wei Chou
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | | | - Dong-Tsamn Lin
- Childhood Cancer Foundation, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsuan-Yu Chen
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Ya-Hsuan Chang
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan.
| | - Yung-Li Yang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
18
|
Steinemann D, Dawidowska M, Russell LJ, Harrison CJ, Göhring G. Genetic alterations in lymphoblastic leukaemia / lymphoma - a practical guide to WHO HAEM5. MED GENET-BERLIN 2024; 36:39-45. [PMID: 38835965 PMCID: PMC11006319 DOI: 10.1515/medgen-2024-2007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
We present a practical guide for analyzing the genetic aspects of lymphoblastic leukaemia/lymphoma according to the 5th edition of the World Health Organization (WHO) classification of haematolymphoid neoplasms (WHO-HAEM5) issued in 2024. The WHO-HAEM5 acknowledges the increasing importance of genetics in the diagnosis of lymphoid neoplasia. Classification is based on the established genetic subtypes according to cell lineage, with precursor cell neoplasms followed by mature malignancies. This guide describes those genetic abnormalities in acute precursor B- and T-cell neoplasms required for risk stratification, and for treatment, providing diagnostic algorithms under the headings of 'essential' and 'desirable' diagnostic criteria.
Collapse
Affiliation(s)
- Doris Steinemann
- Hannover Medical School Department of Human Genetics Hannover Germany
| | - Małgorzata Dawidowska
- Institute of Human Genetics Department of Molecular and Clinical Genetics Poznan Poland
| | - Lisa J Russell
- Newcastle University Centre for Cancer Biosciences Institute Newcastle upon Tyne UK
| | - Christine J Harrison
- Newcastle University Centre for Cancer Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle upon Tyne UK
| | - Gudrun Göhring
- Amedes genetics MVZ wagnerstibbe für Laboratoriumsmedizin, Hämostaseologie, Humangenetik und Mikrobiologie Hannover Germany
| |
Collapse
|
19
|
Popov A, Henze G, Tsaur G, Budanov O, Roumiantseva J, Belevtsev M, Verzhbitskaya T, Movchan L, Lagoyko S, Zharikova L, Olshanskaya Y, Riger T, Valochnik A, Miakova N, Litvinov D, Khlebnikova O, Streneva O, Stolyarova E, Ponomareva N, Novichkova G, Aleinikova O, Fechina L, Karachunskiy A. Flow cytometric minimal residual disease measurement accounting for cytogenetics in children with non-high-risk acute lymphoblastic leukemia treated according to the ALL-MB 2008 protocol. Cancer Med 2024; 13:e7172. [PMID: 38651186 PMCID: PMC11036069 DOI: 10.1002/cam4.7172] [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: 11/17/2023] [Revised: 02/15/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Quantitative measurement of minimal residual disease (MRD) is the "gold standard" for estimating the response to therapy in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Nevertheless, the speed of the MRD response differs for different cytogenetic subgroups. Here we present results of MRD measurement in children with BCP-ALL, in terms of genetic subgroups with relation to clinically defined risk groups. METHODS A total of 485 children with non-high-risk BCP-ALL with available cytogenetic data and MRD studied at the end-of-induction (EOI) by multicolor flow cytometry (MFC) were included. All patients were treated with standard-risk (SR) of intermediate-risk (ImR) regimens of "ALL-MB 2008" reduced-intensity protocol. RESULTS AND DISCUSSION Among all study group patients, 203 were found to have low-risk cytogenetics (ETV6::RUNX1 or high hyperdiploidy), while remaining 282 children were classified in intermediate cytogenetic risk group. For the patients with favorable and intermediate risk cytogenetics, the most significant thresholds for MFC-MRD values were different: 0.03% and 0.04% respectively. Nevertheless, the most meaningful thresholds were different for clinically defined SR and ImR groups. For the SR group, irrespective to presence/absence of favorable genetic lesions, MFC-MRD threshold of 0.1% was the most clinically valuable, although for ImR group the most informative thresholds were different in patients from low-(0.03%) and intermediate (0.01%) cytogenetic risk groups. CONCLUSION Our data show that combining clinical risk factors with MFC-MRD measurement is the most useful tool for risk group stratification of children with BCP-ALL in the reduced-intensity protocols. However, this algorithm can be supplemented with cytogenetic data for part of the ImR group.
Collapse
Affiliation(s)
- Alexander Popov
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Guenter Henze
- Department of Pediatric Oncology HematologyCharité—Universitätsmedizin BerlinBerlinGermany
| | - Grigory Tsaur
- Regional Children's HospitalEkaterinburgRussian Federation
- Research Institute of Medical Cell TechnologiesEkaterinburgRussian Federation
- Ural State Medical UniversityEkaterinburgRussian Federation
| | - Oleg Budanov
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Julia Roumiantseva
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Mikhail Belevtsev
- Republican Scientific and Practical Center for Pediatric OncologyHematology and ImmunologyMinskBelarus
| | - Tatiana Verzhbitskaya
- Regional Children's HospitalEkaterinburgRussian Federation
- Research Institute of Medical Cell TechnologiesEkaterinburgRussian Federation
| | - Liudmila Movchan
- Republican Scientific and Practical Center for Pediatric OncologyHematology and ImmunologyMinskBelarus
| | - Svetlana Lagoyko
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Liudmila Zharikova
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Yulia Olshanskaya
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Tatiana Riger
- Regional Children's HospitalEkaterinburgRussian Federation
| | - Alena Valochnik
- Republican Scientific and Practical Center for Pediatric OncologyHematology and ImmunologyMinskBelarus
| | - Natalia Miakova
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Dmitry Litvinov
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | | | - Olga Streneva
- Regional Children's HospitalEkaterinburgRussian Federation
- Research Institute of Medical Cell TechnologiesEkaterinburgRussian Federation
| | | | - Natalia Ponomareva
- Pirogov Russian National Research Medical UniversityMoscowRussian Federation
| | - Galina Novichkova
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Olga Aleinikova
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| | - Larisa Fechina
- Regional Children's HospitalEkaterinburgRussian Federation
- Research Institute of Medical Cell TechnologiesEkaterinburgRussian Federation
| | - Alexander Karachunskiy
- National Research and Clinical Center for Pediatric Hematology, Oncology and ImmunologyMoscowRussian Federation
| |
Collapse
|
20
|
Mosquera Orgueira A, Krali O, Pérez Míguez C, Peleteiro Raíndo A, Díaz Arias JÁ, González Pérez MS, Pérez Encinas MM, Fernández Sanmartín M, Sinnet D, Heyman M, Lönnerholm G, Norén-Nyström U, Schmiegelow K, Nordlund J. Refining risk prediction in pediatric acute lymphoblastic leukemia through DNA methylation profiling. Clin Epigenetics 2024; 16:49. [PMID: 38549146 PMCID: PMC10976833 DOI: 10.1186/s13148-024-01662-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/16/2024] [Indexed: 04/02/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most prevalent cancer in children, and despite considerable progress in treatment outcomes, relapses still pose significant risks of mortality and long-term complications. To address this challenge, we employed a supervised machine learning technique, specifically random survival forests, to predict the risk of relapse and mortality using array-based DNA methylation data from a cohort of 763 pediatric ALL patients treated in Nordic countries. The relapse risk predictor (RRP) was constructed based on 16 CpG sites, demonstrating c-indexes of 0.667 and 0.677 in the training and test sets, respectively. The mortality risk predictor (MRP), comprising 53 CpG sites, exhibited c-indexes of 0.751 and 0.754 in the training and test sets, respectively. To validate the prognostic value of the predictors, we further analyzed two independent cohorts of Canadian (n = 42) and Nordic (n = 384) ALL patients. The external validation confirmed our findings, with the RRP achieving a c-index of 0.667 in the Canadian cohort, and the RRP and MRP achieving c-indexes of 0.529 and 0.621, respectively, in an independent Nordic cohort. The precision of the RRP and MRP models improved when incorporating traditional risk group data, underscoring the potential for synergistic integration of clinical prognostic factors. The MRP model also enabled the definition of a risk group with high rates of relapse and mortality. Our results demonstrate the potential of DNA methylation as a prognostic factor and a tool to refine risk stratification in pediatric ALL. This may lead to personalized treatment strategies based on epigenetic profiling.
Collapse
Affiliation(s)
- Adrián Mosquera Orgueira
- Department of Hematology, University Hospital of Santiago de Compostela, Compostela, Spain.
- Health Research Institute of Santiago de Compostela, Compostela, Spain.
| | - Olga Krali
- Department of Medical Sciences, Molecular Precision Medicine, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Andrés Peleteiro Raíndo
- Department of Hematology, University Hospital of Santiago de Compostela, Compostela, Spain
- Health Research Institute of Santiago de Compostela, Compostela, Spain
| | - José Ángel Díaz Arias
- Department of Hematology, University Hospital of Santiago de Compostela, Compostela, Spain
- Health Research Institute of Santiago de Compostela, Compostela, Spain
| | - Marta Sonia González Pérez
- Department of Hematology, University Hospital of Santiago de Compostela, Compostela, Spain
- Health Research Institute of Santiago de Compostela, Compostela, Spain
| | - Manuel Mateo Pérez Encinas
- Department of Hematology, University Hospital of Santiago de Compostela, Compostela, Spain
- Health Research Institute of Santiago de Compostela, Compostela, Spain
| | - Manuel Fernández Sanmartín
- Health Research Institute of Santiago de Compostela, Compostela, Spain
- Department of Pediatric Medicine, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Daniel Sinnet
- Research Center, CHU Sainte-Justine, Montréal, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Canada
| | - Mats Heyman
- Childhood Cancer Research Unit, Karolinska Institutet, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
- For the Nordic Society of Pediatric Hematology and Oncology (NOPHO), Stockholm, Sweden
| | - Gudmar Lönnerholm
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
- For the Nordic Society of Pediatric Hematology and Oncology (NOPHO), Stockholm, Sweden
| | - Ulrika Norén-Nyström
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
- For the Nordic Society of Pediatric Hematology and Oncology (NOPHO), Stockholm, Sweden
| | - Kjeld Schmiegelow
- Pediatrics and Adolescent Medicine, Rigshospitalet, and the Medical Faculty, Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- For the Nordic Society of Pediatric Hematology and Oncology (NOPHO), Stockholm, Sweden
| | - Jessica Nordlund
- Department of Medical Sciences, Molecular Precision Medicine, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
21
|
Kubota H, Ueno H, Tasaka K, Isobe T, Saida S, Kato I, Umeda K, Hiwatari M, Hasegawa D, Imamura T, Kakiuchi N, Nannya Y, Ogawa S, Hiramatsu H, Takita J. RNA-seq-based miRNA signature as an independent predictor of relapse in pediatric B-cell acute lymphoblastic leukemia. Blood Adv 2024; 8:1258-1271. [PMID: 38127276 PMCID: PMC10918494 DOI: 10.1182/bloodadvances.2023011583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/21/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
ABSTRACT Aberrant micro-RNA (miRNA) expression profiles have been associated with disease progression and clinical outcome in pediatric cancers. However, few studies have analyzed genome-wide dysregulation of miRNAs and messenger RNAs (mRNAs) in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL). To identify novel prognostic factors, we comprehensively investigated miRNA and mRNA sequencing (miRNA-seq and mRNA-seq) data in pediatric BCP-ALL samples with poor outcome. We analyzed 180 patients, including 43 matched pairs at diagnosis and relapse. Consensus clustering of miRNA expression data revealed a distinct profile characterized by mainly downregulation of miRNAs (referred to as an miR-low cluster [MLC]). The MLC profile was not associated with any known genetic subgroups. Intriguingly, patients classified as MLC had significantly shorter event-free survival (median 21 vs 33 months; log-rank P = 3 ×10-5). Furthermore, this poor prognosis was retained even in hyperdiploid ALL. This poor prognostic MLC profiling was confirmed in the validation cohort. Notably, non-MLC profiling at diagnosis (n = 9 of 23; Fisher exact test, P = .039) often changed into MLC profiling at relapse for the same patient. Integrated analysis of miRNA-seq and mRNA-seq data revealed that the transcriptional profile of MLC was characterized by enrichment of MYC target and oxidative phosphorylation genes, reduced intron retention, and low expression of DICER1. Thus, our miRNA-mRNA integration approach yielded a truly unbiased molecular stratification of pediatric BCP-ALL cases based on a novel prognostic miRNA signature, which may lead to better clinical outcomes.
Collapse
Affiliation(s)
- Hirohito Kubota
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroo Ueno
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiji Tasaka
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoya Isobe
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Hematology, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Satoshi Saida
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Itaru Kato
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mitsuteru Hiwatari
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Pediatrics, School of Medicine, Teikyo University, Tokyo, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Hyogo Prefectural Kobe Children Hospital, Hyogo, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nobuyuki Kakiuchi
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- The Hakubi Center for Advanced Research, Kyoto University, Kyoto, Japan
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Division of Hematopoietic Disease Control, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hidefumi Hiramatsu
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
22
|
Padmakumar A, Thankamony P, Vasudevan JA, Gopinath P, Chandraprabha VR, Devi ARTV, Anitha GRJ, Sreelatha MM, Padmakumar D, Sreedharan H. Double Philadelphia chromosome: a rare and sole abnormality in pediatric B-acute lymphoblastic leukemia. 3 Biotech 2024; 14:75. [PMID: 38371902 PMCID: PMC10866820 DOI: 10.1007/s13205-024-03935-6] [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: 08/19/2022] [Accepted: 12/04/2023] [Indexed: 02/20/2024] Open
Abstract
The present study describes a 7-year-old male child who had attended the Pediatric Oncology Clinic of the Regional Cancer Centre, Thiruvananthapuram, Kerala, India, and was pathologically confirmed to have B-Acute Lymphoblastic Leukemia (B-ALL). Conventional cytogenetics analysis at diagnosis showed the presence of a double Philadelphia chromosome and the karyotype of the case was 47, XY, t(9;22)(q34;q11.2), + der(22)t(9;22). FISH, done as a molecular confirmation of the translocation, t(9;22)(q34;q11.2), and this case showed an additional fusion signal that confirms the presence of double Ph. As far as we are aware, this represents the initial and only occurrence of an abnormality report regarding the double Philadelphia chromosome in pediatric B-ALL within India. The double Philadelphia chromosome in B-ALL has a very poor prognosis despite aggressive treatment with chemotherapy. This study reveals the importance of conventional and molecular cytogenetic analysis in risk stratification and prognosis prediction of pediatric B-ALL. The risk stratification based on the conventional and molecular cytogenetic analysis may be taken into consideration for deciding the treatment strategy for each patient.
Collapse
Affiliation(s)
- Amritha Padmakumar
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, University of Kerala, Thiruvananthapuram, India
| | | | | | - Preethi Gopinath
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, University of Kerala, Thiruvananthapuram, India
| | - Vineetha Radhakrishnan Chandraprabha
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, University of Kerala, Thiruvananthapuram, India
| | - Akhila Raj Thampirajan Vimala Devi
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, University of Kerala, Thiruvananthapuram, India
| | - Geetha Raj John Anitha
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, University of Kerala, Thiruvananthapuram, India
| | - Mahitha Mohanan Sreelatha
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, University of Kerala, Thiruvananthapuram, India
| | - Devipriya Padmakumar
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, University of Kerala, Thiruvananthapuram, India
| | - Hariharan Sreedharan
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, University of Kerala, Thiruvananthapuram, India
| |
Collapse
|
23
|
Kolekar P, Balagopal V, Dong L, Liu Y, Foy S, Tran Q, Mulder H, Huskey AL, Plyler E, Liang Z, Ma J, Nakitandwe J, Gu J, Namwanje M, Maciaszek J, Payne-Turner D, Mallampati S, Wang L, Easton J, Klco JM, Ma X. SJPedPanel: A pan-cancer gene panel for childhood malignancies. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.11.27.23299068. [PMID: 38076942 PMCID: PMC10705664 DOI: 10.1101/2023.11.27.23299068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Background Large scale genomics projects have identified driver alterations for most childhood cancers that provide reliable biomarkers for clinical diagnosis and disease monitoring using targeted sequencing. However, there is lack of a comprehensive panel that matches the list of known driver genes. Here we fill this gap by developing SJPedPanel for childhood cancers. Results SJPedPanel covers 5,275 coding exons of 357 driver genes, 297 introns frequently involved in rearrangements that generate fusion oncoproteins, commonly amplified/deleted regions (e.g., MYCN for neuroblastoma, CDKN2A and PAX5 for B-/T-ALL, and SMARCB1 for AT/RT), and 7,590 polymorphism sites for interrogating tumors with aneuploidy, such as hyperdiploid and hypodiploid B-ALL or 17q gain neuroblastoma. We used driver alterations reported from an established real-time clinical genomics cohort (n=253) to validate this gene panel. Among the 485 pathogenic variants reported, our panel covered 417 variants (86%). For 90 rearrangements responsible for oncogenic fusions, our panel covered 74 events (82%). We re-sequenced 113 previously characterized clinical specimens at an average depth of 2,500X using SJPedPanel and recovered 354 (91%) of the 389 reported pathogenic variants. We then investigated the power of this panel in detecting mutations from specimens with low tumor purity (as low as 0.1%) using cell line-based dilution experiments and discovered that this gene panel enabled us to detect ∼80% variants with allele fraction of 0.2%, while the detection rate decreases to ∼50% when the allele fraction is 0.1%. We finally demonstrate its utility in disease monitoring on clinical specimens collected from AML patients in morphologic remission. Conclusions SJPedPanel enables the detection of clinically relevant genetic alterations including rearrangements responsible for subtype-defining fusions for childhood cancers by targeted sequencing of ∼0.15% of human genome. It will enhance the analysis of specimens with low tumor burdens for cancer monitoring and early detection.
Collapse
|
24
|
Lee SHR. Toward a Comprehensive One-Stop Shop for Somatic Genomic Profiling in Childhood Acute Lymphoblastic Leukemia. J Mol Diagn 2024; 26:2-4. [PMID: 37972852 DOI: 10.1016/j.jmoldx.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Affiliation(s)
- Shawn H R Lee
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
25
|
Hu Z, Kovach AE, Yellapantula V, Ostrow D, Doan A, Ji J, Schmidt RJ, Gu Z, Bhojwani D, Raca G. Transcriptome Sequencing Allows Comprehensive Genomic Characterization of Pediatric B-Acute Lymphoblastic Leukemia in an Academic Clinical Laboratory. J Mol Diagn 2024; 26:49-60. [PMID: 37981088 PMCID: PMC10773144 DOI: 10.1016/j.jmoldx.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/16/2023] [Accepted: 09/28/2023] [Indexed: 11/21/2023] Open
Abstract
Studies have shown the power of transcriptome sequencing [RNA sequencing (RNA-Seq)] in identifying known and novel oncogenic drivers and molecular subtypes of B-acute lymphoblastic leukemia (B-ALL). The current study investigated whether the clinically validated RNA-Seq assay, coupled with a custom analysis pipeline, could be used for a comprehensive B-ALL classification. Following comprehensive clinical testing, RNA-Seq was performed on 76 retrospective B-ALL cases, 28 of which had known and 48 had undetermined subtype. Subtypes were accurately identified in all 28 known cases, and in 38 of 48 unknown cases (79%). The subtypes of the unknown cases included the following: PAX5alt (n = 12), DUX4-rearranged (n = 6), Philadelphia chromosome-like (n = 5), low hyperdiploid (n = 4), ETV6::RUNX1-like (n = 3), MEF2D-rearranged (n = 2), PAX5 P80R (n = 2), ZEB2/CEBP (n = 1), NUTM1-rearranged (n = 1), ZNF384-rearranged (n = 1), and TCF3::PBX1 (n = 1). In 15 of 38 cases (39%), classification based on expression profile was corroborated by detection of subtype-defining oncogenic drivers missed by clinical testing. RNA-Seq analysis also detected large copy number abnormalities, oncogenic hot-spot sequence variants, and intragenic IKZF1 deletions. This pilot study confirms the feasibility of implementing an RNA-Seq workflow for clinical diagnosis of molecular subtypes in pediatric B-ALL, reinforcing that RNA-Seq represents a promising global genomic assay for this heterogeneous leukemia.
Collapse
Affiliation(s)
- Zunsong Hu
- Department of Computational and Quantitative Medicine and Systems Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Alexandra E Kovach
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Venkata Yellapantula
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Dejerianne Ostrow
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Andrew Doan
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California
| | - Jianling Ji
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Ryan J Schmidt
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Zhaohui Gu
- Department of Computational and Quantitative Medicine and Systems Biology, Beckman Research Institute of City of Hope, Duarte, California.
| | - Deepa Bhojwani
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California
| | - Gordana Raca
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California.
| |
Collapse
|
26
|
Monovich AC, Gurumurthy A, Ryan RJH. The Diverse Roles of ETV6 Alterations in B-Lymphoblastic Leukemia and Other Hematopoietic Cancers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:291-320. [PMID: 39017849 DOI: 10.1007/978-3-031-62731-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Genetic alterations of the repressive ETS family transcription factor gene ETV6 are recurrent in several categories of hematopoietic malignancy, including subsets of B-cell and T-cell acute lymphoblastic leukemias (B-ALL and T-ALL), myeloid neoplasms, and mature B-cell lymphomas. ETV6 is essential for adult hematopoietic stem cells (HSCs), contributes to specific functions of some mature immune cells, and plays a key role in thrombopoiesis as demonstrated by familial ETV6 mutations associated with thrombocytopenia and predisposition to hematopoietic cancers, particularly B-ALL. ETV6 appears to have a tumor suppressor role in several hematopoietic lineages, as demonstrated by recurrent somatic loss-of-function (LoF) and putative dominant-negative alterations in leukemias and lymphomas. ETV6 rearrangements contribute to recurrent fusion oncogenes such as the B-ALL-associated transcription factor (TF) fusions ETV6::RUNX1 and PAX5::ETV6, rare drivers such as ETV6::NCOA6, and a spectrum of tyrosine kinase gene fusions encoding hyperactive signaling proteins that self-associate via the ETV6 N-terminal pointed domain. Another subset of recurrent rearrangements involving the ETV6 gene locus appear to function primarily to drive overexpression of the partner gene. This review surveys what is known about the biochemical and genome regulatory properties of ETV6 as well as our current understanding of how alterations in these functions contribute to hematopoietic and nonhematopoietic cancers.
Collapse
Affiliation(s)
- Alexander C Monovich
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Aishwarya Gurumurthy
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Russell J H Ryan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
27
|
Pieters R, Mullighan CG, Hunger SP. Advancing Diagnostics and Therapy to Reach Universal Cure in Childhood ALL. J Clin Oncol 2023; 41:5579-5591. [PMID: 37820294 PMCID: PMC10730082 DOI: 10.1200/jco.23.01286] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/24/2023] [Accepted: 08/10/2023] [Indexed: 10/13/2023] Open
Abstract
Systemic combination chemotherapy and intrathecal chemotherapy markedly increased the survival rate of children with ALL. In the past two decades, the use of minimal (measurable) residual disease (MRD) measurements early in therapy improved risk group stratification with subsequent treatment intensifications for patients at high risk of relapse, and enabled a reduction of treatment for low-risk patients. The recent development of more sensitive MRD technologies may further affect risk stratification. Molecular genetic profiling has led to the discovery of many new subtypes and their driver genetic alterations. This increased our understanding of the biological basis of ALL, improved risk classification, and enabled implementation of precision medicine. In the past decade, immunotherapies, including bispecific antibodies, antibody-drug conjugates, and cellular therapies directed against surface proteins, led to more effective and less toxic therapies, replacing intensive chemotherapy courses and allogeneic stem-cell transplantation in patients with relapsed and refractory ALL, and are now being tested in newly diagnosed patients. It has taken 50-60 years to increase the cure rate in childhood ALL from 0% to 90% by stepwise improvements in chemotherapy. This review provides an overview of how the developments over the past 10-15 years mentioned above have significantly changed the diagnostic and treatment approach in ALL, and discusses how the integrated use of molecular and immunotherapeutic insights will very likely direct efforts to cure those children with ALL who are not cured today, and improve the quality of life for survivors who should have decades of life ahead. Future efforts must focus on making effective, yet very expensive, new technologies and therapies available to children with ALL worldwide.
Collapse
Affiliation(s)
- Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Charles G. Mullighan
- Department of Pathology and Hematological Malignancies Program, Comprehensive Cancer Center, St Jude Children's Research Hospital, Memphis, TN
| | - Stephen P. Hunger
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
28
|
Lee SH, Ashcraft E, Yang W, Roberts KG, Gocho Y, Rowland L, Inaba H, Karol SE, Jeha S, Crews KR, Mullighan CG, Relling MV, Evans WE, Cheng C, Yang JJ, Pui CH. Prognostic and Pharmacotypic Heterogeneity of Hyperdiploidy in Childhood ALL. J Clin Oncol 2023; 41:5422-5432. [PMID: 37729596 PMCID: PMC10852380 DOI: 10.1200/jco.23.00880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/29/2023] [Accepted: 07/26/2023] [Indexed: 09/22/2023] Open
Abstract
PURPOSE High hyperdiploidy, the largest and favorable subtype of childhood ALL, exhibits significant biological and prognostic heterogeneity. However, factors contributing to the varied treatment response and the optimal definition of hyperdiploidy remain uncertain. METHODS We analyzed outcomes of patients treated on two consecutive frontline ALL protocols, using six different definitions of hyperdiploidy: chromosome number 51-67 (Chr51-67); DNA index (DI; DI1.16-1.6); United Kingdom ALL study group low-risk hyperdiploid, either trisomy of chromosomes 17 and 18 or +17 or +18 in the absence of +5 and +20; single trisomy of chromosome 18; double trisomy of chromosomes 4 and 10; and triple trisomy (TT) of chromosomes 4, 10, and 17. Additionally, we characterized ALL ex vivo pharmacotypes across eight main cytotoxic drugs. RESULTS Among 1,096 patients analyzed, 915 had B-ALL and 634 had pharmacotyping performed. In univariate analysis, TT emerged as the most favorable criterion for event-free survival (EFS; 10-year EFS, 97.3% v 86.8%; P = .0003) and cumulative incidence of relapse (CIR; 10-year CIR, 1.4% v 8.8%; P = .002) compared with the remaining B-ALL. In multivariable analysis, accounting for patient numbers using the akaike information criterion (AIC), DI1.16-1.6 was the most favorable criterion, exhibiting the best AIC for both EFS (hazard ratio [HR], 0.45; 95% CI, 0.23 to 0.88) and CIR (HR, 0.45; 95% CI, 0.21 to 0.99). Hyperdiploidy and subgroups with favorable prognoses exhibited notable sensitivities to asparaginase and mercaptopurine. Specifically, asparaginase sensitivity was associated with trisomy of chromosomes 16 and 17, whereas mercaptopurine sensitivity was linked to gains of chromosomes 14 and 17. CONCLUSION Among different definitions of hyperdiploid ALL, DI is optimal based on independent prognostic impact and also the large proportion of low-risk patients identified. Hyperdiploid ALL exhibited particular sensitivities to asparaginase and mercaptopurine, with chromosome-specific associations.
Collapse
Affiliation(s)
- Shawn H.R. Lee
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Emily Ashcraft
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN
| | - Wenjian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
| | - Kathryn G. Roberts
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN
| | - Yoshihiro Gocho
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
| | - Lauren Rowland
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
| | - Hiroto Inaba
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Seth E. Karol
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Sima Jeha
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Kristine R. Crews
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
| | | | - Mary V. Relling
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
| | - William E. Evans
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
| | - Cheng Cheng
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN
| | - Jun J. Yang
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Ching-Hon Pui
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
29
|
Pelayo R. A new hope for early T cell precursor acute lymphoblastic leukemia therapy based on STAT5+ leukemic stem cell targeting. J Leukoc Biol 2023; 114:381-383. [PMID: 37607260 DOI: 10.1093/jleuko/qiad094] [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: 06/12/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/24/2023] Open
Abstract
Leukemia stem cells are known to drive tumor progression, drug resistance, microenvironmental shift, and relapse, which would make them a perfect therapeutic target. However, their phenotypic and functional similarity to their normal counterparts leaves limited road maps for their selective elimination. Tremblay et al. recently unraveled the fundamental role of overactivated pSTAT5 as a functional marker of early T cell precursor acute lymphoblastic leukemia stem cells driving leukemic progression and highlighted its potential use as a therapeutic target to prevent fatal outcomes.
Collapse
Affiliation(s)
- Rosana Pelayo
- Unidad de Educación e Investigación, Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Oriente, Km 4.5 Carretera Atlixco-Metepec, 74360 Puebla, Mexico
| |
Collapse
|
30
|
Tripathi S, Shirnekhi HK, Gorman SD, Chandra B, Baggett DW, Park CG, Somjee R, Lang B, Hosseini SMH, Pioso BJ, Li Y, Iacobucci I, Gao Q, Edmonson MN, Rice SV, Zhou X, Bollinger J, Mitrea DM, White MR, McGrail DJ, Jarosz DF, Yi SS, Babu MM, Mullighan CG, Zhang J, Sahni N, Kriwacki RW. Defining the condensate landscape of fusion oncoproteins. Nat Commun 2023; 14:6008. [PMID: 37770423 PMCID: PMC10539325 DOI: 10.1038/s41467-023-41655-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
Fusion oncoproteins (FOs) arise from chromosomal translocations in ~17% of cancers and are often oncogenic drivers. Although some FOs can promote oncogenesis by undergoing liquid-liquid phase separation (LLPS) to form aberrant biomolecular condensates, the generality of this phenomenon is unknown. We explored this question by testing 166 FOs in HeLa cells and found that 58% formed condensates. The condensate-forming FOs displayed physicochemical features distinct from those of condensate-negative FOs and segregated into distinct feature-based groups that aligned with their sub-cellular localization and biological function. Using Machine Learning, we developed a predictor of FO condensation behavior, and discovered that 67% of ~3000 additional FOs likely form condensates, with 35% of those predicted to function by altering gene expression. 47% of the predicted condensate-negative FOs were associated with cell signaling functions, suggesting a functional dichotomy between condensate-positive and -negative FOs. Our Datasets and reagents are rich resources to interrogate FO condensation in the future.
Collapse
Affiliation(s)
- Swarnendu Tripathi
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hazheen K Shirnekhi
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Scott D Gorman
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Arrakis Therapeutics, 830 Winter St, Waltham, MA, 02451, USA
| | - Bappaditya Chandra
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David W Baggett
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cheon-Gil Park
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ramiz Somjee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Rhodes College, Memphis, TN, USA
- Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Benjamin Lang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center of Excellence for Data-Driven Discovery, Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Seyed Mohammad Hadi Hosseini
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center of Excellence for Data-Driven Discovery, Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brittany J Pioso
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yongsheng Li
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Qingsong Gao
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael N Edmonson
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen V Rice
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - John Bollinger
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Diana M Mitrea
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Dewpoint Therapeutics, 451 D Street, Suite 104, Boston, MA, 02210, USA
| | - Michael R White
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- IDEXX Laboratories, Inc., One IDEXX Drive, Westbrook, ME, 04092, USA
| | - Daniel J McGrail
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Daniel F Jarosz
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - S Stephen Yi
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Biomedical Engineering, and Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, USA
| | - M Madan Babu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center of Excellence for Data-Driven Discovery, Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA
| | - Richard W Kriwacki
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center, Memphis, TN, USA.
| |
Collapse
|
31
|
Gao Q, Ryan SL, Iacobucci I, Ghate PS, Cranston RE, Schwab C, Elsayed AH, Shi L, Pounds S, Lei S, Baviskar P, Pei D, Cheng C, Bashton M, Sinclair P, Bentley DR, Ross MT, Kingsbury Z, James T, Roberts KG, Devidas M, Fan Y, Chen W, Chang TC, Wu G, Carroll A, Heerema N, Valentine V, Valentine M, Yang W, Yang JJ, Moorman AV, Harrison CJ, Mullighan CG. The genomic landscape of acute lymphoblastic leukemia with intrachromosomal amplification of chromosome 21. Blood 2023; 142:711-723. [PMID: 37216686 PMCID: PMC10460677 DOI: 10.1182/blood.2022019094] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Intrachromosomal amplification of chromosome 21 defines a subtype of high-risk childhood acute lymphoblastic leukemia (iAMP21-ALL) characterized by copy number changes and complex rearrangements of chromosome 21. The genomic basis of iAMP21-ALL and the pathogenic role of the region of amplification of chromosome 21 to leukemogenesis remains incompletely understood. In this study, using integrated whole genome and transcriptome sequencing of 124 patients with iAMP21-ALL, including rare cases arising in the context of constitutional chromosomal aberrations, we identified subgroups of iAMP21-ALL based on the patterns of copy number alteration and structural variation. This large data set enabled formal delineation of a 7.8 Mb common region of amplification harboring 71 genes, 43 of which were differentially expressed compared with non-iAMP21-ALL ones, including multiple genes implicated in the pathogenesis of acute leukemia (CHAF1B, DYRK1A, ERG, HMGN1, and RUNX1). Using multimodal single-cell genomic profiling, including single-cell whole genome sequencing of 2 cases, we documented clonal heterogeneity and genomic evolution, demonstrating that the acquisition of the iAMP21 chromosome is an early event that may undergo progressive amplification during disease ontogeny. We show that UV-mutational signatures and high mutation load are characteristic secondary genetic features. Although the genomic alterations of chromosome 21 are variable, these integrated genomic analyses and demonstration of an extended common minimal region of amplification broaden the definition of iAMP21-ALL for more precise diagnosis using cytogenetic or genomic methods to inform clinical management.
Collapse
Affiliation(s)
- Qingsong Gao
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Sarra L. Ryan
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, United Kingdom
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Pankaj S. Ghate
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ruth E. Cranston
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, United Kingdom
| | - Claire Schwab
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, United Kingdom
| | - Abdelrahman H. Elsayed
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Lei Shi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Shaohua Lei
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Deqing Pei
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Matthew Bashton
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, United Kingdom
| | - Paul Sinclair
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, United Kingdom
| | - David R. Bentley
- Illumina Cambridge, Ltd, Illumina Centre, Great Abingdon, Cambridge, United Kingdom
| | - Mark T. Ross
- Illumina Cambridge, Ltd, Illumina Centre, Great Abingdon, Cambridge, United Kingdom
| | - Zoya Kingsbury
- Illumina Cambridge, Ltd, Illumina Centre, Great Abingdon, Cambridge, United Kingdom
| | - Terena James
- Illumina Cambridge, Ltd, Illumina Centre, Great Abingdon, Cambridge, United Kingdom
| | - Kathryn G. Roberts
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Wenan Chen
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Andrew Carroll
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Nyla Heerema
- Department of Pathology, The Ohio State University, Columbus, OH
| | - Virginia Valentine
- Cytogenetics Shared Resource, St. Jude Children’s Research Hospital, Memphis, TN
| | - Marcus Valentine
- Cytogenetics Shared Resource, St. Jude Children’s Research Hospital, Memphis, TN
| | - Wenjian Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jun J. Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Anthony V. Moorman
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, United Kingdom
| | - Christine J. Harrison
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, United Kingdom
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
32
|
Yilmaz B, Koc A, Dogru O, Tufan Tas B, Senay RE. The results of the modified St Jude Total Therapy XV Protocol in the treatment of low- and middle-income children with acute lymphoblastic leukemia. Leuk Lymphoma 2023; 64:1304-1314. [PMID: 37165575 DOI: 10.1080/10428194.2023.2205976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 05/12/2023]
Abstract
The St Jude Total Therapy Study XV was the first clinical trial to prospectively use minimal residual disease levels during and after remission induction therapy to guide risk-directed treatment. We used the Total Therapy XV protocol with minimal modification in treating 115 newly diagnosed pediatric acute lymphoblastic leukemia patients from low- and middle-income groups from January 2011 to December 2017. The mean age at diagnosis was 5.97 ± 3.96 years. The median follow-up period was 88 months. Three (2.6%) patients had bone marrow relapse, and one (0.87%) had an isolated central nervous system relapse. Nineteen of the patients (16.52%) died due to infection-related complications, three (2.61%) died due to progressive disease, and one (0.87%) died due to hematopoietic stem cell transplant complications. Five-year overall survival was 80%, and event-free survival was 78.3%. Our results showed that the Total XV treatment protocol could be used successfully in patients with ALL from low- and middle-income populations. However, infection-related deaths remain a significant problem.
Collapse
Affiliation(s)
- Baris Yilmaz
- Pediatric Hematology and Oncology, SB Marmara University Medical School Education and Research Hospital, Istanbul, Türkiye
| | - Ahmet Koc
- Pediatric Hematology and Oncology, SB Marmara University Medical School Education and Research Hospital, Istanbul, Türkiye
| | - Omer Dogru
- Pediatric Hematology and Oncology, SB Marmara University Medical School Education and Research Hospital, Istanbul, Türkiye
| | - Burcu Tufan Tas
- Pediatric Hematology and Oncology, SB Marmara University Medical School Education and Research Hospital, Istanbul, Türkiye
| | - Rabia Emel Senay
- Pediatric Hematology and Oncology, SB Marmara University Medical School Education and Research Hospital, Istanbul, Türkiye
| |
Collapse
|
33
|
Talleur AC, Pui CH, Karol SE. What is Next in Pediatric B-cell Precursor Acute Lymphoblastic Leukemia. LYMPHATICS 2023; 1:34-44. [PMID: 38269058 PMCID: PMC10804398 DOI: 10.3390/lymphatics1010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Cure rates now exceed 90% in many contemporary trials for children with B-cell acute lymphoblastic leukemia (ALL). However, treatment remains suboptimal and therapy is toxic for all patients. New treatment options potentially offer the chance to reduce both treatment resistance and toxicity. Here, we review recent advances in ALL diagnostics, chemotherapy, and immunotherapy. In addition to describing recently published results, we also attempt to project the impact of these new developments into the future to imagine what B-ALL therapy may look like in the next few years.
Collapse
Affiliation(s)
- Aimee C Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
34
|
Dekker SE, Rea D, Cayuela JM, Arnhardt I, Leonard J, Heuser M. Using Measurable Residual Disease to Optimize Management of AML, ALL, and Chronic Myeloid Leukemia. Am Soc Clin Oncol Educ Book 2023; 43:e390010. [PMID: 37311155 DOI: 10.1200/edbk_390010] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In this review, we discuss the use of measurable residual disease (MRD) in AML, ALL, and chronic myeloid leukemia (CML). Our aims were to review the different methodologies for MRD assessment; describe the clinical relevance and medical decision making on the basis of MRD; compare and contrast the usage of MRD across AML, ALL, and CML; and discuss what patients need to know about MRD as it relates to their disease status and treatment. Finally, we discuss ongoing challenges and future directions with the goal of optimizing MRD usage in leukemia management.
Collapse
Affiliation(s)
- Simone E Dekker
- Department of Medicine, Oregon Health & Science University, Portland, OR
| | - Delphine Rea
- France Intergroupe des Leucémies Myéloïdes chroniques FiLMC, Hôpital Saint-Louis APHP, Paris, France
- Service d'Hématologie Adulte, Hôpital Saint-Louis APHP, Paris, France
| | - Jean-Michel Cayuela
- France Intergroupe des Leucémies Myéloïdes chroniques FiLMC, Hôpital Saint-Louis APHP, Paris, France
- Laboratoire de Biologie Moléculaire, Hôpital Saint-Louis APHP, Paris, France
| | - Isabell Arnhardt
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Jessica Leonard
- Division of Hematology-Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- Comprehensive Cancer Center Lower Saxony, Hannover, Germany
| |
Collapse
|
35
|
Panuciak K, Nowicka E, Mastalerczyk A, Zawitkowska J, Niedźwiecki M, Lejman M. Overview on Aneuploidy in Childhood B-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2023; 24:ijms24108764. [PMID: 37240110 DOI: 10.3390/ijms24108764] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Recent years have brought significant progress in the treatment of B-cell acute lymphoblastic leukemia (ALL). This was influenced by both the improved schemes of conventionally used therapy, as well as the development of new forms of treatment. As a consequence, 5-year survival rates have increased and now exceed 90% in pediatric patients. For this reason, it would seem that everything has already been explored in the context of ALL. However, delving into its pathogenesis at the molecular level shows that there are many variations that still need to be analyzed in more detail. One of them is aneuploidy, which is among the most common genetic changes in B-cell ALL. It includes both hyperdiploidy and hypodiploidy. Knowledge of the genetic background is important already at the time of diagnosis, because the first of these forms of aneuploidy is characterized by a good prognosis, in contrast to the second, which is in favor of an unfavorable course. In our work, we will focus on summarizing the current state of knowledge on aneuploidy, along with an indication of all the consequences that may be correlated with it in the context of the treatment of patients with B-cell ALL.
Collapse
Affiliation(s)
- Kinga Panuciak
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Emilia Nowicka
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Angelika Mastalerczyk
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland
| | - Maciej Niedźwiecki
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
36
|
Gu Z, Hu Z, Jia Z, Liu J, Mao A, Han H. MD-ALL: an Integrative Platform for Molecular Diagnosis of B-cell Acute Lymphoblastic Leukemia. RESEARCH SQUARE 2023:rs.3.rs-2798895. [PMID: 37090504 PMCID: PMC10120769 DOI: 10.21203/rs.3.rs-2798895/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) consists of dozens of subtypes defined by distinct gene expression profiles (GEPs) and various genetic lesions. With the application of transcriptome sequencing (RNA-seq), multiple novel subtypes have been identified, which lead to an advanced B-ALL classification and risk-stratification system. However, the complexity of analyzing RNA-seq data for B-ALL classification hinders the implementation of the new B-ALL taxonomy. Here, we introduce MD-ALL (Molecular Diagnosis of ALL), a user-friendly platform featuring sensitive and accurate B-ALL classification based on GEPs and sentinel genetic alterations. In this study, we systematically analyzed 2,955 B-ALL RNA-seq samples and generated a reference dataset representing all the reported B-ALL subtypes. Using multiple machine learning algorithms, we identified the feature genes and then established highly accurate models for B-ALL classification using either bulk or single-cell RNA-seq data. Importantly, this platform integrates the key genetic lesions, including sequence mutations, large-scale copy number variations, and gene rearrangements, to perform comprehensive and definitive B-ALL classification. Through validation in a hold-out cohort of 974 samples, our models demonstrated superior performance for B-ALL classification compared with alternative tools. In summary, MD-ALL is a user-friendly B-ALL classification platform designed to enable integrative, accurate, and comprehensive B-ALL subtype classification.
Collapse
|
37
|
Imai C, Sato A, Hiwatari M, Shimomura Y, Hori T, Suenobu S, Imamura T, Hara J, Hasegawa D, Takahashi H, Moriya K, Katayama S, Tomizawa D, Moritake H, Taga T, Horibe K, Koh K, Manabe A, Okamoto Y. Outcomes following induction failure in Japanese children with acute lymphoblastic leukemia. Int J Hematol 2023:10.1007/s12185-023-03600-3. [PMID: 37037958 DOI: 10.1007/s12185-023-03600-3] [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: 01/23/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/12/2023]
Abstract
The characteristics and prognosis of Japanese children with acute lymphoblastic leukemia (ALL) who fail to achieve complete remission after remission induction chemotherapy (i.e., experience induction failure) are poorly understood. Therefore, we retrospectively analyzed data of patients enrolled in Japanese clinical trials for newly diagnosed ALL between 1996 and 2009. Among 4956 participants, 89 (1.8%) experienced induction failure. With a 6.0-year median follow-up, the 5-year overall survival rate of the entire cohort was 43.0% ± 5.5%. Survival rates did not differ between patients with B-cell precursor ALL (BCP-ALL) and T-cell ALL (T-ALL). In multivariate analysis, day 15 M3 marrow (bone marrow blast count ≥ 25%) was significantly correlated with poorer survival in the whole or BCP-ALL cohorts. In T-ALL, age < 6 years was significantly associated with poor survival. However, due to the small sample size, this correlation must be further investigated. Most T-ALL and BCR-ABL-positive BCP-ALL patients underwent allogeneic stem cell transplantation (allo-SCT). Survival rates did not differ between BCR-ABL-negative BCP-ALL patients who did and did not undergo allo-SCT, possibly due to the inclusion of lower-risk patients in the latter group. In conclusion, the induction failure rate and survival after diagnosis of induction failure in our study were comparable to previously reported figures.
Collapse
Affiliation(s)
- Chihaya Imai
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-Dori, Chuou-Ku, Niigata City, Niigata, 951-8510, Japan.
| | - Atsushi Sato
- Department of Hematology and Oncology, Miyagi Children's Hospital, Miyagi, Japan
| | - Mitsuteru Hiwatari
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Yasuto Shimomura
- Department of Pediatrics, Aichi Medical University, Aichi, Japan
| | - Toshinori Hori
- Department of Pediatrics, Aichi Medical University, Aichi, Japan
| | | | - Toshihiko Imamura
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junichi Hara
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Hiroyuki Takahashi
- Department of Pediatrics, Toho University Omori Medical Center, Tokyo, Japan
| | - Kunihiko Moriya
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Saori Katayama
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Daisuke Tomizawa
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Hiroshi Moritake
- Department of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Keizo Horibe
- Clinical Research Center, Nagoya Medical Center, Nagoya, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Yasuhiro Okamoto
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
38
|
Davis K, Sheikh T, Aggarwal N. Emerging molecular subtypes and therapies in acute lymphoblastic leukemia. Semin Diagn Pathol 2023; 40:202-215. [PMID: 37120350 DOI: 10.1053/j.semdp.2023.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 05/01/2023]
Abstract
Tremendous strides have been made in the molecular and cytogenetic classification of acute lymphoblastic leukemia based on gene expression profiling data, leading to an expansion of entities in the recent International Consensus Classification (ICC) of myeloid neoplasms and acute leukemias and 2022 WHO Classification of Tumours: Haematolymphoid Tumors, 5th edition. This increased diagnostic and therapeutic complexity can be overwhelming, and this review compares nomenclature differences between the ICC and WHO 5th edition publications, compiles key features of each entity, and provides a diagnostic algorithmic approach. In covering B-lymphoblastic leukemia (B-ALL), we divided the entities into established (those present in the revised 4th edition WHO) and novel (those added to either the ICC or WHO 5th edition) groups. The established B-ALL entities include B-ALL with BCR::ABL1 fusion, BCR::ABL1-like features, KMT2A rearrangement, ETV6::RUNX1 rearrangement, high hyperdiploidy, hypodiploidy (focusing on near haploid and low hypodiploid), IGH::IL3 rearrangement, TCF3::PBX1 rearrangement, and iAMP21. The novel B-ALL entities include B-ALL with MYC rearrangement; DUX4 rearrangement; MEF2D rearrangement; ZNF384 or ZNF362 rearrangement, NUTM1 rearrangement; HLF rearrangement; UBTF::ATXN7L3/PAN3,CDX2; mutated IKZF1 N159Y; mutated PAX5 P80R; ETV6::RUNX1-like features; PAX5 alteration; mutated ZEB2 (p.H1038R)/IGH::CEBPE; ZNF384 rearranged-like; KMT2A-rearranged-like; and CRLF2 rearrangement (non-Ph-like). Classification of T-ALL is complex with some variability in how the subtypes are defined in recent literature. It was classified as early T-precursor lymphoblastic leukemia/lymphoma and T-ALL, NOS in the WHO revised 4th edition and WHO 5th edition. The ICC added an entity into early T-cell precursor ALL, BCL11B-activated, and also added provisional entities subclassified based on transcription factor families that are aberrantly activated.
Collapse
Affiliation(s)
- Katelynn Davis
- Department of Hematopathology, School of Medicine and UPMC, University of Pittsburgh, USA
| | | | - Nidhi Aggarwal
- Department of Hematopathology, School of Medicine and UPMC, University of Pittsburgh, USA.
| |
Collapse
|
39
|
Schwab C, Cranston RE, Ryan SL, Butler E, Winterman E, Hawking Z, Bashton M, Enshaei A, Russell LJ, Kingsbury Z, Peden JF, Barretta E, Murray J, Gibson J, Hinchliffe AC, Bain R, Vora A, Bentley DR, Ross MT, Moorman AV, Harrison CJ. Integrative genomic analysis of childhood acute lymphoblastic leukaemia lacking a genetic biomarker in the UKALL2003 clinical trial. Leukemia 2023; 37:529-538. [PMID: 36550215 PMCID: PMC9991913 DOI: 10.1038/s41375-022-01799-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Incorporating genetics into risk-stratification for treatment of childhood B-progenitor acute lymphoblastic leukaemia (B-ALL) has contributed significantly to improved survival. In about 30% B-ALL (B-other-ALL) without well-established chromosomal changes, new genetic subtypes have recently emerged, yet their true prognostic relevance largely remains unclear. We integrated next generation sequencing (NGS): whole genome sequencing (WGS) (n = 157) and bespoke targeted NGS (t-NGS) (n = 175) (overlap n = 36), with existing genetic annotation in a representative cohort of 351 B-other-ALL patients from the childhood ALL trail, UKALL2003. PAX5alt was most frequently observed (n = 91), whereas PAX5 P80R mutations (n = 11) defined a distinct PAX5 subtype. DUX4-r subtype (n = 80) was defined by DUX4 rearrangements and/or ERG deletions. These patients had a low relapse rate and excellent survival. ETV6::RUNX1-like subtype (n = 21) was characterised by multiple abnormalities of ETV6 and IKZF1, with no reported relapses or deaths, indicating their excellent prognosis in this trial. An inferior outcome for patients with ABL-class fusions (n = 25) was confirmed. Integration of NGS into genomic profiling of B-other-ALL within a single childhood ALL trial, UKALL2003, has shown the added clinical value of NGS-based approaches, through improved accuracy in detection and classification into the range of risk stratifying genetic subtypes, while validating their prognostic significance.
Collapse
Affiliation(s)
- Claire Schwab
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Ruth E Cranston
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Sarra L Ryan
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Ellie Butler
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Emily Winterman
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Zoe Hawking
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Matthew Bashton
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Amir Enshaei
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Lisa J Russell
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Zoya Kingsbury
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - John F Peden
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - Emilio Barretta
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - James Murray
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Jude Gibson
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Andrew C Hinchliffe
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Robert Bain
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Ajay Vora
- Department of Haematology, Great Ormond Street Hospital, London, UK
| | - David R Bentley
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - Mark T Ross
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Christine J Harrison
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK.
| |
Collapse
|
40
|
Kovach AE, Raca G. Modern Classification and Management of Pediatric B-cell Leukemia and Lymphoma. Surg Pathol Clin 2023; 16:249-266. [PMID: 37149359 DOI: 10.1016/j.path.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Although pediatric hematopathology overlaps with that of adults, certain forms of leukemia and lymphoma, and many types of reactive conditions affecting the bone marrow and lymph nodes, are unique to children. As part of this series focused on lymphomas, this article (1) details the novel subtypes of lymphoblastic leukemia seen primarily in children and described since the 2017 World Health Organization classification and (2) discusses unique concepts in pediatric hematopathology, including nomenclature changes and evaluation of surgical margins in selected lymphomas.
Collapse
Affiliation(s)
- Alexandra E Kovach
- Division of Laboratory Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA; Clinical Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA.
| | - Gordana Raca
- Clinical Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA; Division of Genomic Medicine, Department of Pathology and Laboratory Medicine, Center for Personalized Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
41
|
Ryan SL, Peden JF, Kingsbury Z, Schwab CJ, James T, Polonen P, Mijuskovic M, Becq J, Yim R, Cranston RE, Hedges DJ, Roberts KG, Mullighan CG, Vora A, Russell LJ, Bain R, Moorman AV, Bentley DR, Harrison CJ, Ross MT. Whole genome sequencing provides comprehensive genetic testing in childhood B-cell acute lymphoblastic leukaemia. Leukemia 2023; 37:518-528. [PMID: 36658389 PMCID: PMC9991920 DOI: 10.1038/s41375-022-01806-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/20/2023]
Abstract
Childhood B-cell acute lymphoblastic leukaemia (B-ALL) is characterised by recurrent genetic abnormalities that drive risk-directed treatment strategies. Using current techniques, accurate detection of such aberrations can be challenging, due to the rapidly expanding list of key genetic abnormalities. Whole genome sequencing (WGS) has the potential to improve genetic testing, but requires comprehensive validation. We performed WGS on 210 childhood B-ALL samples annotated with clinical and genetic data. We devised a molecular classification system to subtype these patients based on identification of key genetic changes in tumour-normal and tumour-only analyses. This approach detected 294 subtype-defining genetic abnormalities in 96% (202/210) patients. Novel genetic variants, including fusions involving genes in the MAP kinase pathway, were identified. WGS results were concordant with standard-of-care methods and whole transcriptome sequencing (WTS). We expanded the catalogue of genetic profiles that reliably classify PAX5alt and ETV6::RUNX1-like subtypes. Our novel bioinformatic pipeline improved detection of DUX4 rearrangements (DUX4-r): a good-risk B-ALL subtype with high survival rates. Overall, we have validated that WGS provides a standalone, reliable genetic test to detect all subtype-defining genetic abnormalities in B-ALL, accurately classifying patients for the risk-directed treatment stratification, while simultaneously performing as a research tool to identify novel disease biomarkers.
Collapse
Affiliation(s)
- Sarra L Ryan
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - John F Peden
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - Zoya Kingsbury
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - Claire J Schwab
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Terena James
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - Petri Polonen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Jenn Becq
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - Richard Yim
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Ruth E Cranston
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Dale J Hedges
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kathryn G Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ajay Vora
- Department of Haematology, Great Ormond Street Hospital, London, UK
| | - Lisa J Russell
- Biosciences Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Robert Bain
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Anthony V Moorman
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - David R Bentley
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK
| | - Christine J Harrison
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle upon Tyne, UK.
| | - Mark T Ross
- Illumina Cambridge Ltd., Granta Park, Great Abington, Cambridge, UK.
| |
Collapse
|
42
|
Cremaschi A, Yang W, De Iorio M, Evans WE, Yang JJ, Rosner GL. Bayesian modelling of response to therapy and drug-sensitivity in acute lymphoblastic leukemia. RESEARCH SQUARE 2023:rs.3.rs-2542277. [PMID: 36865272 PMCID: PMC9980194 DOI: 10.21203/rs.3.rs-2542277/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is a heterogeneous haematologic malignancy involving the abnormal proliferation of immature lymphocytes and accounts for most paediatric cancer cases. The management of ALL in children has seen great improvement in the last decades thanks to greater understanding of the disease leading to improved treatment strategies evidenced through clinical trials. Common therapy regimens involve a first course of chemotherapy (induction phase), followed by treatment with a combination of anti-leukemia drugs. A measure of the efficacy early in the course of therapy is the presence of minimal residual disease (MRD). MRD quantifies residual tumor cells and indicates the effiectiveness of the treatment over the course of therapy. MRD positivity is defined for values of MRD greater than 0.01%, yielding left-censored MRD observations. We propose a Bayesian model to study the relationship between patient features (leukemia subtype, baseline characteristics, and drug sensitivity profile) and MRD observed at two time points during the induction phase. Specifically, we model the observed MRD values via an auto-regressive model, accounting for left-censoring of the data and for the fact that some patients are already in remission after the first stage of induction therapy. Patient characteristics are included in the model via linear regression terms. In particular, patient-specific drug sensitivity based on ex vivo assays of patient samples is exploited to identify groups of subjects with similar profiles. We include this information as a covariate in the model for MRD. We adopt horseshoe priors for the regression coefficients to perform variable selection to identify important covariates. We fit the proposed approach to data from three prospective paediatric ALL clinical trials carried out at the St. Jude Children's Research Hospital. Our results highlight that drug sensitivity profiles and leukemic subtypes play an important role in the response to induction therapy as measured by serial MRD measures.
Collapse
Affiliation(s)
- Andrea Cremaschi
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Singapore
| | - Wenjian Yang
- St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Maria De Iorio
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Statistical Science, University College London, UK
| | | | - Jun J. Yang
- St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Gary L. Rosner
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Chiaretti S, Taherinasab A, Della Starza I, Canichella M, Ansuinelli M, De Propris MS, Messina M, Spinelli O, Santoro A, De Novi LA, Cardinali D, Schipani M, Arena V, Bassan R, Guarini A, Foà R. ZNF384 rearrangement is the most frequent genetic lesion in adult PH-negative and Ph-like-negative B-other acute lymphoblastic leukemia. Biological and clinical findings. Leuk Lymphoma 2023; 64:483-486. [PMID: 36533589 DOI: 10.1080/10428194.2022.2148217] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Sabina Chiaretti
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Akram Taherinasab
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy.,Fondazione GIMEMA Onlus, Rome, Italy
| | - Martina Canichella
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Michela Ansuinelli
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Maria Stefania De Propris
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | | | - Orietta Spinelli
- Haematology and Bone Marrow Transplant Unit, ASST-Papa Giovanni XXIII, Bergamo
| | - Alessandra Santoro
- Department of Hemato Oncology, AOR "VillaSofia-Cervello", Palermo, Italy
| | - Lucia Anna De Novi
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Deborah Cardinali
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Mattia Schipani
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | | | - Renato Bassan
- Division of Hematology, Ospedale dell'Angelo, Mestre, Venezia, Italy
| | - Anna Guarini
- Department of Molecular Medicine, "Sapienza" University of Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
44
|
Kato M. Recent progress in pediatric lymphoblastic leukemia. Int J Hematol 2023; 117:155-161. [PMID: 36456860 DOI: 10.1007/s12185-022-03501-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
The probability of long-term survival for children with lymphoblastic leukemia has improved dramatically over recent decades, mainly owing to advances in genomic analysis techniques, which have improved our understanding of the nature of leukemic cells and prognostic prediction based on the evaluation of precise treatment response. Risk-adjusted chemotherapy based on these advances has simultaneously reduced relapse rates and minimized complications. In addition, recent genomic analyses have deepened our understanding of the pathogenesis of leukemia and revealed the involvement of germline variations in the clinical course of leukemia treatment. Additionally, advances in minimal residual disease assays and the introduction of immunotherapy are expected to further improve therapeutic analyses. Further advances in clinical and translational research are anticipated to improve survival to 100% in a healthy state.
Collapse
Affiliation(s)
- Motohiro Kato
- Department of Pediatrics, The University of Tokyo, 7-3-1, Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
45
|
Mangum DS, Bishop JD, Zhou Y, Cheng C, Karol SE, Rubnitz JE, Ribeiro RC, Yang JJ, Mullighan CG, Jeha S, Pui CH, Inaba H. Characterisation of children and adolescents with acute lymphoblastic leukaemia who presented without peripheral blood blasts at diagnosis. Br J Haematol 2023; 200:338-343. [PMID: 36352514 PMCID: PMC9852218 DOI: 10.1111/bjh.18552] [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: 08/30/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022]
Abstract
Of 1003 children with acute lymphoblastic leukaemia (ALL), 147 (14.7%) presented without peripheral blood blasts (PBB). While absence of PBB was not independently associated with survival outcomes when compared to those with PBB, patients without PBB had distinct genetic and clinical characteristics. Notably, we identified a novel genotype-phenotype relationship, in that the patients without PBB had a significantly higher incidence of hyperdiploid B-ALL, accounting for almost half of all patients without PBB (46.9% vs. 22.7%, p < 0.001). Further, absence of PBB was associated with decreased rates of leukaemia involvement of the central nervous system (p < 0.001).
Collapse
Affiliation(s)
- D. Spencer Mangum
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Division of Hematology/Oncology, Department of Pediatrics, University of Utah and Primary Children’s Hospital, Salt Lake City, UT, USA
| | - Johnathon D. Bishop
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children’s Hospital, Memphis, Tennessee, USA
| | - Yinmei Zhou
- Department of Bioinformatics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Cheng Cheng
- Department of Bioinformatics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Seth E. Karol
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeffrey E. Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Raul C. Ribeiro
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sima Jeha
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
46
|
Iacobucci I, Witkowski MT, Mullighan CG. Single-cell analysis of acute lymphoblastic and lineage-ambiguous leukemia: approaches and molecular insights. Blood 2023; 141:356-368. [PMID: 35926109 PMCID: PMC10023733 DOI: 10.1182/blood.2022016954] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/13/2022] [Accepted: 07/23/2022] [Indexed: 01/31/2023] Open
Abstract
Despite recent progress in identifying the genetic drivers of acute lymphoblastic leukemia (ALL), prognosis remains poor for those individuals who experience disease recurrence. Moreover, acute leukemias of ambiguous lineage lack a biologically informed framework to guide classification and therapy. These needs have driven the adoption of multiple complementary single-cell sequencing approaches to explore key issues in the biology of these leukemias, including cell of origin, developmental hierarchy and ontogeny, and the molecular heterogeneity driving pathogenesis, progression, and therapeutic responsiveness. There are multiple single-cell techniques for profiling a specific modality, including RNA, DNA, chromatin accessibility and methylation; and an expanding range of approaches for simultaneous analysis of multiple modalities. Single-cell sequencing approaches have also enabled characterization of cell-intrinsic and -extrinsic features of ALL biology. In this review we describe these approaches and highlight the extensive heterogeneity that underpins ALL gene expression, cellular differentiation, and clonal architecture throughout disease pathogenesis and treatment resistance. In addition, we discuss the importance of the dynamic interactions that occur between leukemia cells and the nonleukemia microenvironment. We discuss potential opportunities and limitations of single-cell sequencing for the study of ALL biology and treatment responsiveness.
Collapse
Affiliation(s)
- Ilaria Iacobucci
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN
| | - Matthew T. Witkowski
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Charles G. Mullighan
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN
- Hematological Malignancies Program, St Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
47
|
Lee SHR, Yang W, Gocho Y, John A, Rowland L, Smart B, Williams H, Maxwell D, Hunt J, Yang W, Crews KR, Roberts KG, Jeha S, Cheng C, Karol SE, Relling MV, Rosner GL, Inaba H, Mullighan CG, Pui CH, Evans WE, Yang JJ. Pharmacotypes across the genomic landscape of pediatric acute lymphoblastic leukemia and impact on treatment response. Nat Med 2023; 29:170-179. [PMID: 36604538 PMCID: PMC9873558 DOI: 10.1038/s41591-022-02112-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/28/2022] [Indexed: 01/07/2023]
Abstract
Contemporary chemotherapy for childhood acute lymphoblastic leukemia (ALL) is risk-adapted based on clinical features, leukemia genomics and minimal residual disease (MRD); however, the pharmacological basis of these prognostic variables remains unclear. Analyzing samples from 805 children with newly diagnosed ALL from three consecutive clinical trials, we determined the ex vivo sensitivity of primary leukemia cells to 18 therapeutic agents across 23 molecular subtypes defined by leukemia genomics. There was wide variability in drug response, with favorable ALL subtypes exhibiting the greatest sensitivity to L-asparaginase and glucocorticoids. Leukemia sensitivity to these two agents was highly associated with MRD although with distinct patterns and only in B cell ALL. We identified six patient clusters based on ALL pharmacotypes, which were associated with event-free survival, even after adjusting for MRD. Pharmacotyping identified a T cell ALL subset with a poor prognosis that was sensitive to targeted agents, pointing to alternative therapeutic strategies. Our study comprehensively described the pharmacological heterogeneity of ALL, highlighting opportunities for further individualizing therapy for this most common childhood cancer.
Collapse
Affiliation(s)
- Shawn H. R. Lee
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA ,grid.412106.00000 0004 0621 9599Khoo Teck Puat–National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wenjian Yang
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Yoshihiro Gocho
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - August John
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Lauren Rowland
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Brandon Smart
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Hannah Williams
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Dylan Maxwell
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Jeremy Hunt
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Wentao Yang
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Kristine R. Crews
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Kathryn G. Roberts
- grid.240871.80000 0001 0224 711XDepartment of Pathology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Sima Jeha
- grid.240871.80000 0001 0224 711XDepartment of Oncology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Cheng Cheng
- grid.240871.80000 0001 0224 711XDepartment of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Seth E. Karol
- grid.240871.80000 0001 0224 711XDepartment of Oncology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Mary V. Relling
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Gary L. Rosner
- grid.280502.d0000 0000 8741 3625Quantitative Sciences, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD USA
| | - Hiroto Inaba
- grid.240871.80000 0001 0224 711XDepartment of Oncology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Charles G. Mullighan
- grid.240871.80000 0001 0224 711XDepartment of Pathology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Ching-Hon Pui
- grid.240871.80000 0001 0224 711XDepartment of Oncology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - William E. Evans
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Jun J. Yang
- grid.240871.80000 0001 0224 711XDepartment of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA ,grid.240871.80000 0001 0224 711XDepartment of Oncology, St. Jude Children’s Research Hospital, Memphis, TN USA
| |
Collapse
|
48
|
Tran TH, Tasian SK. Clinical screening for Ph-like ALL and the developing role of TKIs. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:594-602. [PMID: 36485164 PMCID: PMC9821133 DOI: 10.1182/hematology.2022000357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is a common subtype of B-lineage acute lymphoblastic leukemia (B-ALL) with increasing frequency across the age spectrum. Characterized by a kinase-activated gene expression profile and driven by a variety of genetic alterations involving cytokine receptors and kinases, Ph-like ALL is associated with high rates of residual disease and relapse in patients treated with conventional chemotherapy. In this case-based review, we describe the biology of the 2 major ABL-class and JAK pathway genetic subtypes of Ph-like ALL, discuss current diagnostic testing methodologies, and highlight targeted inhibitor and chemo/immunotherapy approaches under clinical investigation in children, adolescents, and adults with these high-risk leukemias.
Collapse
Affiliation(s)
- Thai Hoa Tran
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
49
|
Haas OA, Borkhardt A. Hyperdiploidy: the longest known, most prevalent, and most enigmatic form of acute lymphoblastic leukemia in children. Leukemia 2022; 36:2769-2783. [PMID: 36266323 PMCID: PMC9712104 DOI: 10.1038/s41375-022-01720-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022]
Abstract
Hyperdiploidy is the largest genetic entity B-cell precursor acute lymphoblastic leukemia in children. The diagnostic hallmark of its two variants that will be discussed in detail herein is a chromosome count between 52 and 67, respectively. The classical HD form consists of heterozygous di-, tri-, and tetrasomies, whereas the nonclassical one (usually viewed as "duplicated hyperhaploid") contains only disomies and tetrasomies. Despite their apparently different clinical behavior, we show that these two sub-forms can in principle be produced by the same chromosomal maldistribution mechanism. Moreover, their respective array, gene expression, and mutation patterns also indicate that they are biologically more similar than hitherto appreciated. Even though in-depth analyses of the genomic intricacies of classical HD leukemias are indispensable for the elucidation of the disease process, the ensuing results play at present surprisingly little role in treatment stratification, a fact that can be attributed to the overall good prognoses and low relapse rates of the concerned patients and, consequently, their excellent treatment outcome. Irrespective of this underutilization, however, the detailed genetic characterization of HD leukemias may, especially in planned treatment reduction trials, eventually become important for further treatment stratification, patient management, and the clinical elucidation of outcome data. It should therefore become an integral part of all upcoming treatment studies.
Collapse
Affiliation(s)
- Oskar A Haas
- St. Anna Children's Hospital, Pediatric Clinic, Medical University, Vienna, Austria.
- Labdia Labordiagnostik, Vienna, Austria.
| | - Arndt Borkhardt
- Department for Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
- German Cancer Consortium (DKTK), partnering site Essen/Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
50
|
Closa A, Reixachs-Solé M, Fuentes-Fayos AC, Hayer K, Melero J, Adriaanse FRS, Bos R, Torres-Diz M, Hunger S, Roberts K, Mullighan C, Stam R, Thomas-Tikhonenko A, Castaño J, Luque R, Eyras E. A convergent malignant phenotype in B-cell acute lymphoblastic leukemia involving the splicing factor SRRM1. NAR Cancer 2022; 4:zcac041. [PMID: 36518527 PMCID: PMC9732526 DOI: 10.1093/narcan/zcac041] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/09/2022] [Accepted: 11/25/2022] [Indexed: 11/07/2024] Open
Abstract
A significant proportion of infant B-cell acute lymphoblastic leukemia (B-ALL) patients remains with a dismal prognosis due to yet undetermined mechanisms. We performed a comprehensive multicohort analysis of gene expression, gene fusions, and RNA splicing alterations to uncover molecular signatures potentially linked to the observed poor outcome. We identified 87 fusions with significant allele frequency across patients and shared functional impacts, suggesting common mechanisms across fusions. We further identified a gene expression signature that predicts high risk independently of the gene fusion background and includes the upregulation of the splicing factor SRRM1. Experiments in B-ALL cell lines provided further evidence for the role of SRRM1 on cell survival, proliferation, and invasion. Supplementary analysis revealed that SRRM1 potentially modulates splicing events associated with poor outcomes through protein-protein interactions with other splicing factors. Our findings reveal a potential convergent mechanism of aberrant RNA processing that sustains a malignant phenotype independently of the underlying gene fusion and that could potentially complement current clinical strategies in infant B-ALL.
Collapse
Affiliation(s)
- Adria Closa
- The Shine-Dalgarno Centre for RNA Innovation, John Curtin School of Medical Research, Australian National University, Canberra, Australia
- Centre for Computational Biomedical Sciences, John Curtin School of Medical Research, Australian National University, Canberra, Australia
- EMBL Australia Partner Laboratory Network at the Australian National University, Canberra, Australia
| | - Marina Reixachs-Solé
- The Shine-Dalgarno Centre for RNA Innovation, John Curtin School of Medical Research, Australian National University, Canberra, Australia
- Centre for Computational Biomedical Sciences, John Curtin School of Medical Research, Australian National University, Canberra, Australia
- EMBL Australia Partner Laboratory Network at the Australian National University, Canberra, Australia
| | - Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba (UCO), Cordoba, Spain
- Reina Sofía University Hospital, Cordoba, Spain
| | - Katharina E Hayer
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Juan L Melero
- The Shine-Dalgarno Centre for RNA Innovation, John Curtin School of Medical Research, Australian National University, Canberra, Australia
- Centre for Computational Biomedical Sciences, John Curtin School of Medical Research, Australian National University, Canberra, Australia
- EMBL Australia Partner Laboratory Network at the Australian National University, Canberra, Australia
| | | | - Romy S Bos
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Manuel Torres-Diz
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Stephen P Hunger
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Kathryn G Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, USA
| | - Ronald W Stam
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba (UCO), Cordoba, Spain
- Reina Sofía University Hospital, Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba (UCO), Cordoba, Spain
- Reina Sofía University Hospital, Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Eduardo Eyras
- The Shine-Dalgarno Centre for RNA Innovation, John Curtin School of Medical Research, Australian National University, Canberra, Australia
- Centre for Computational Biomedical Sciences, John Curtin School of Medical Research, Australian National University, Canberra, Australia
- EMBL Australia Partner Laboratory Network at the Australian National University, Canberra, Australia
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|