1
|
Doculara L, Evans K, Gooding JJ, Bayat N, Lock RB. Patient-Specific Circulating Tumor DNA for Monitoring Response to Menin Inhibitor Treatment in Preclinical Models of Infant Leukemia. Cancers (Basel) 2024; 16:3990. [PMID: 39682177 DOI: 10.3390/cancers16233990] [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: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND In infant KMT2A (MLL1)-rearranged (MLL-r) acute lymphoblastic leukemia (ALL), early relapse and treatment response are currently monitored through invasive repeated bone marrow (BM) biopsies. Circulating tumor DNA (ctDNA) in peripheral blood (PB) provides a minimally invasive alternative, allowing for more frequent disease monitoring. However, a poor understanding of ctDNA dynamics has hampered its clinical translation. We explored the predictive value of ctDNA for detecting minimal/measurable residual disease (MRD) and drug response in a patient-derived xenograft (PDX) model of infant MLL-r ALL. METHODS Immune-deficient mice engrafted with three MLL-r ALL PDXs were monitored for ctDNA levels before and after treatment with the menin inhibitor SNDX-50469. RESULTS The amount of ctDNA detected strongly correlated with leukemia burden during initial engraftment prior to drug treatment. However, following SNDX-50469 treatment, the leukemic burden assessed by either PB leukemia cells through flow cytometry or ctDNA levels through droplet digital polymerase chain reaction (ddPCR) was discrepant. This divergence could be attributed to the persistence of leukemia cells in the spleen and BM, highlighting the ability of ctDNA to reflect disease dynamics in key leukemia infiltration sites. CONCLUSIONS Notably, ctDNA analysis proved to be a superior predictor of MRD compared to PB assessment alone, especially in instances of low disease burden. These findings highlight the potential of ctDNA as a sensitive biomarker for monitoring treatment response and detecting MRD in infant MLL-r ALL.
Collapse
Affiliation(s)
- Louise Doculara
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Kathryn Evans
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - J Justin Gooding
- School of Chemistry, UNSW Sydney, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Narges Bayat
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
2
|
Janssen FW, Lak NSM, Janda CY, Kester LA, Meister MT, Merks JHM, van den Heuvel-Eibrink MM, van Noesel MM, Zsiros J, Tytgat GAM, Looijenga LHJ. A comprehensive overview of liquid biopsy applications in pediatric solid tumors. NPJ Precis Oncol 2024; 8:172. [PMID: 39097671 PMCID: PMC11297996 DOI: 10.1038/s41698-024-00657-z] [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: 02/20/2024] [Accepted: 07/15/2024] [Indexed: 08/05/2024] Open
Abstract
Liquid biopsies are emerging as an alternative source for pediatric cancer biomarkers with potential applications during all stages of patient care, from diagnosis to long-term follow-up. While developments within this field are reported, these mainly focus on dedicated items such as a specific liquid biopsy matrix, analyte, and/or single tumor type. To the best of our knowledge, a comprehensive overview is lacking. Here, we review the current state of liquid biopsy research for the most common non-central nervous system pediatric solid tumors. These include neuroblastoma, renal tumors, germ cell tumors, osteosarcoma, Ewing sarcoma, rhabdomyosarcoma and other soft tissue sarcomas, and liver tumors. Within this selection, we discuss the most important or recent studies involving liquid biopsy-based biomarkers, anticipated clinical applications, and the current challenges for success. Furthermore, we provide an overview of liquid biopsy-based biomarker publication output for each tumor type based on a comprehensive literature search between 1989 and 2023. Per study identified, we list the relevant liquid biopsy-based biomarkers, matrices (e.g., peripheral blood, bone marrow, or cerebrospinal fluid), analytes (e.g., circulating cell-free and tumor DNA, microRNAs, and circulating tumor cells), methods (e.g., digital droplet PCR and next-generation sequencing), the involved pediatric patient cohort, and proposed applications. As such, we identified 344 unique publications. Taken together, while the liquid biopsy field in pediatric oncology is still behind adult oncology, potentially relevant publications have increased over the last decade. Importantly, steps towards clinical implementation are rapidly gaining ground, notably through validation of liquid biopsy-based biomarkers in pediatric clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Michael T Meister
- Princess Máxima Center, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Johannes H M Merks
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center, Utrecht, the Netherlands
- Wilhelmina Children's Hospital-Division of CHILDHEALTH, University Medical Center Utrech, University of Utrecht, Utrecht, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | | | - Godelieve A M Tytgat
- Princess Máxima Center, Utrecht, the Netherlands
- Department of Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Leendert H J Looijenga
- Princess Máxima Center, Utrecht, the Netherlands.
- Department of Pathology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Biglari N, Soltani-Zangbar MS, Mohammadian J, Mehdizadeh A, Abbasi K. ctDNA as a novel and promising approach for cancer diagnosis: a focus on hepatocellular carcinoma. EXCLI JOURNAL 2023; 22:752-780. [PMID: 37720239 PMCID: PMC10502204 DOI: 10.17179/excli2023-6277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 07/26/2023] [Indexed: 09/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent forms of cancer worldwide. Therefore, it is essential to diagnose and treat HCC patients promptly. As a novel discovery, circulating tumor DNA (ctDNA) can be used to analyze the tumor type and the cancer location. Additionally, ctDNA assists the cancer stage determination, which enables medical professionals to provide patients with the most appropriate treatment. This review will discuss the HCC-related mutated genes diagnosed by ctDNA. In addition, we will introduce the different and the most appropriate ctDNA diagnosis approaches based on the facilities.
Collapse
Affiliation(s)
- Negin Biglari
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Sadegh Soltani-Zangbar
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jamal Mohammadian
- School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Abbasi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Buenache N, Sánchez-delaCruz A, Cuenca I, Giménez A, Moreno L, Martínez-López J, Rosa-Rosa JM. Identification of Immunoglobulin Gene Rearrangement Biomarkers in Multiple Myeloma through cfDNA-Based Liquid Biopsy Using tchDNA-Seq. Cancers (Basel) 2023; 15:cancers15112911. [PMID: 37296872 DOI: 10.3390/cancers15112911] [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: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the clonal proliferation of pathogenic CD138+ plasma cells (PPCs) in bone marrow (BM). Recent years have seen a significant increase in the treatment options for MM; however, most patients who achieve complete the response ultimately relapse. The earlier detection of tumor-related clonal DNA would thus be very beneficial for patients with MM and would enable timely therapeutic interventions to improve outcomes. Liquid biopsy of "cell-free DNA" (cfDNA) as a minimally invasive approach might be more effective than BM aspiration not only for the diagnosis but also for the detection of early recurrence. Most studies thus far have addressed the comparative quantification of patient-specific biomarkers in cfDNA with PPCs and BM samples, which have shown good correlations. However, there are limitations to this approach, such as the difficulty in obtaining enough circulating free tumor DNA to achieve sufficient sensitivity for the assessment of minimal residual disease. Herein, we summarize current data on methodologies to characterize MM, and we present evidence that targeted capture hybridization DNA sequencing (tchDNA-Seq) can provide robust biomarkers in cfDNA, including immunoglobulin (IG) rearrangements. We also show that detection can be improved by prior purification of the cfDNA. Overall, liquid biopsies of cfDNA to monitor IG rearrangements have the potential to provide important diagnostic, prognostic, and predictive information in patients with MM.
Collapse
Affiliation(s)
- Natalia Buenache
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Andrea Sánchez-delaCruz
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Isabel Cuenca
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Alicia Giménez
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Laura Moreno
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Joaquín Martínez-López
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
- Department of Translational Haematology, Haematology Service, Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28034 Madrid, Spain
| | - Juan Manuel Rosa-Rosa
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28034 Madrid, Spain
| |
Collapse
|
5
|
Sato S, Nakamura Y, Oki E, Yoshino T. Molecular Residual Disease-guided Adjuvant Treatment in Resected Colorectal Cancer: Focus on CIRCULATE-Japan. Clin Colorectal Cancer 2023; 22:53-58. [PMID: 36567192 DOI: 10.1016/j.clcc.2022.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
The body of evidence supporting the utility of the detection of molecular residual disease (MRD) in resected colorectal cancer (CRC) using circulating tumor DNA (ctDNA) analysis is rapidly growing. Furthermore, this evidence provides the rationale for escalation and de-escalation adjuvant chemotherapy (ACT) strategies using ctDNA MRD analysis. This has led to various randomized clinical trials, and CIRCULATE-Japan is one of the largest of these trial platforms. In this review, we provide an overview of the potential utility of ctDNA-based MRD detection for escalation and de-escalation ACT approaches. Furthermore, we highlight the feasibility using ctDNA clearance as a surrogate endpoint for ACT trials in patients with resected CRC, based on findings of the CIRCULATE-Japan project.
Collapse
Affiliation(s)
- Seiya Sato
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan; International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan; Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan; Division for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
6
|
van Zogchel LMJ, Lak NSM, Gelineau NU, Sergeeva I, Stelloo E, Swennenhuis J, Feitsma H, van Min M, Splinter E, Bleijs M, Groot Koerkamp M, Breunis W, Meister MT, Kholossy WH, Holstege FCP, Molenaar JJ, de Leng WWJ, Stutterheim J, van der Schoot CE, Tytgat GAM. Targeted locus amplification to develop robust patient-specific assays for liquid biopsies in pediatric solid tumors. Front Oncol 2023; 13:1124737. [PMID: 37152023 PMCID: PMC10157037 DOI: 10.3389/fonc.2023.1124737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/27/2023] [Indexed: 05/09/2023] Open
Abstract
Background Liquid biopsies combine minimally invasive sample collection with sensitive detection of residual disease. Pediatric malignancies harbor tumor-driving copy number alterations or fusion genes, rather than recurrent point mutations. These regions contain tumor-specific DNA breakpoint sequences. We investigated the feasibility to use these breakpoints to design patient-specific markers to detect tumor-derived cell-free DNA (cfDNA) in plasma from patients with pediatric solid tumors. Materials and methods Regions of interest (ROI) were identified through standard clinical diagnostic pipelines, using SNP array for CNAs, and FISH or RT-qPCR for fusion genes. Using targeted locus amplification (TLA) on tumor organoids grown from tumor material or targeted locus capture (TLC) on FFPE material, ROI-specific primers and probes were designed, which were used to design droplet digital PCR (ddPCR) assays. cfDNA from patient plasma at diagnosis and during therapy was analyzed. Results TLA was performed on material from 2 rhabdomyosarcoma, 1 Ewing sarcoma and 3 neuroblastoma. FFPE-TLC was performed on 8 neuroblastoma tumors. For all patients, at least one patient-specific ddPCR was successfully designed and in all diagnostic plasma samples the patient-specific markers were detected. In the rhabdomyosarcoma and Ewing sarcoma patients, all samples after start of therapy were negative. In neuroblastoma patients, presence of patient-specific markers in cfDNA tracked tumor burden, decreasing during induction therapy, disappearing at complete remission and re-appearing at relapse. Conclusion We demonstrate the feasibility to determine tumor-specific breakpoints using TLA/TLC in different pediatric solid tumors and use these for analysis of cfDNA from plasma. Considering the high prevalence of CNAs and fusion genes in pediatric solid tumors, this approach holds great promise and deserves further study in a larger cohort with standardized plasma sampling protocols.
Collapse
Affiliation(s)
- Lieke M. J. van Zogchel
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Sanquin Research and Landsteiner Laboratory of the AMC‐ University of Amsterdam, Department of Experimental Immunohematology, Amsterdam, Netherlands
| | - Nathalie S. M. Lak
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Sanquin Research and Landsteiner Laboratory of the AMC‐ University of Amsterdam, Department of Experimental Immunohematology, Amsterdam, Netherlands
| | - Nina U. Gelineau
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Sanquin Research and Landsteiner Laboratory of the AMC‐ University of Amsterdam, Department of Experimental Immunohematology, Amsterdam, Netherlands
| | | | | | | | | | | | | | - Margit Bleijs
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | | | - Willemijn Breunis
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- University Children’s Hospital Zürich, Zürich, Switzerland
| | - Michael Torsten Meister
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | | | - Frank C. P. Holstege
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Center (UMC) Utrecht and Utrecht University, Utrecht, Netherlands
| | - Jan J. Molenaar
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | - Wendy W. J. de Leng
- Department of Pathology, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Janine Stutterheim
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | - C. Ellen van der Schoot
- Sanquin Research and Landsteiner Laboratory of the AMC‐ University of Amsterdam, Department of Experimental Immunohematology, Amsterdam, Netherlands
| | - Godelieve A. M. Tytgat
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- *Correspondence: Godelieve A. M. Tytgat,
| |
Collapse
|
7
|
Venn NC, Huang L, Hovorková L, Muskovic W, Wong M, Law T, Heatley SL, Khaw SL, Revesz T, Dalla Pozza L, Shaw PJ, Fraser C, Moore AS, Cross S, Bendak K, Norris MD, Henderson MJ, White DL, Cowley MJ, Trahair TN, Zuna J, Sutton R. Measurable residual disease analysis in paediatric acute lymphoblastic leukaemia patients with ABL-class fusions. Br J Cancer 2022; 127:908-915. [PMID: 35650277 PMCID: PMC9427854 DOI: 10.1038/s41416-022-01806-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 03/12/2022] [Accepted: 03/25/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND ABL-class fusions including NUP214-ABL1 and EBF1-PDGFRB occur in high risk acute lymphoblastic leukaemia (ALL) with gene expression patterns similar to BCR-ABL-positive ALL. Our aim was to evaluate new DNA-based measurable residual disease (MRD) tests detecting these fusions and IKZF1-deletions in comparison with conventional immunoglobulin/T-cell receptor (Ig/TCR) markers. METHODS Precise genomic breakpoints were defined from targeted or whole genome next generation sequencing for ABL-fusions and BCR-ABL1. Quantitative PCR assays were designed and used to re-measure MRD in remission bone marrow samples previously tested using Ig/TCR markers. All MRD testing complied with EuroMRD guidelines. RESULTS ABL-class patients had 46% 5year event-free survival and 79% 5year overall survival. All had sensitive fusion tests giving high concordance between Ig/TCR and ABL-class fusion results (21 patients, n = 257 samples, r2 = 0.9786, P < 0.0001) and Ig/TCR and IKZF1-deletion results (9 patients, n = 143 samples, r2 = 0.9661, P < 0.0001). In contrast, in BCR-ABL1 patients, Ig/TCR and BCR-ABL1 tests were discordant in 32% (40 patients, n = 346 samples, r2 = 0.4703, P < 0.0001) and IKZF1-deletion results were closer to Ig/TCR (25 patients, n = 176, r2 = 0.8631, P < 0.0001). CONCLUSIONS MRD monitoring based on patient-specific assays detecting gene fusions or recurrent assays for IKZF1-deletions is feasible and provides good alternatives to Ig/TCR tests to monitor MRD in ABL-class ALL.
Collapse
Affiliation(s)
- Nicola C Venn
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Libby Huang
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Lenka Hovorková
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- CLIP-Childhood Leukaemia Investigation Prague, Prague, Czech Republic
| | - Walter Muskovic
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Marie Wong
- Computational Biology, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Tamara Law
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Susan L Heatley
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Seong Lin Khaw
- Children's Cancer Centre, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Tom Revesz
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
- Department of Clinical Haematology and Oncology, Women's and Children's Hospital, Adelaide, SA, Australia
| | - Luciano Dalla Pozza
- Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Peter J Shaw
- Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Chris Fraser
- Blood and Bone Marrow Transplant Program, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Andrew S Moore
- Paediatric Oncology, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Siobhan Cross
- Children's Haematology/Oncology Centre Christchurch Hospital, Christchurch, New Zealand
| | - Katerina Bendak
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Murray D Norris
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
| | - Michelle J Henderson
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
| | - Deborah L White
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Mark J Cowley
- Computational Biology, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
| | - Toby N Trahair
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Jan Zuna
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- CLIP-Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Rosemary Sutton
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia.
- School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|