TCF3 breakpoints of TCF3-PBX1 (patients 1a 5a) and TCF3-HLF (patients 6a 9a and11a) translocations.

Similar documents
Standard risk ALL (and its exceptions

ETP - Acute Lymphoblastic Leukaemia

MRD in ALL: Correct interpretation in clinical practice. Deepak Bansal Prof., Pediatric Hematology-Oncology unit PGIMER, Chandigarh

BCR ABL1 like ALL: molekuliniai mechanizmai ir klinikinė reikšmė. IKAROS delecija: molekulinė biologija, prognostinė reikšmė. ASH 2015 naujienos

Risk Stratification in Childhood Leukemia

Objectives. Morphology and IHC. Flow and Cyto FISH. Testing for Heme Malignancies 3/20/2013

Nature Immunology: doi: /ni.3412

SUPPLEMENTARY INFORMATION

Molecular Markers in Acute Leukemia. Dr Muhd Zanapiah Zakaria Hospital Ampang

Acute Lymphoblastic Leukemia (ALL) Ryan Mattison, MD University of Wisconsin March 2, 2010

Elisabeth Koller 3rd Medical Dept., Center for Hematology and Oncology, Hanusch Hospital, Vienna, Austria

PAX5-JAK2 fusion in acute lymphoblastic leukaemia. Dr Andrew Baldi Royal Children s Hospital 24 February 2017 Melbourne

Relapse Cytogenetics Overview of ALLR3 genetics Introduction to the IntReALL trial. Anthony V Moorman Leukaemia Research Cytogenetics Group

A pediatric patient with acute leukemia of ambiguous lineage with a NUP98-NSD1 rearrangement SH

Molecular Markers. Marcie Riches, MD, MS Associate Professor University of North Carolina Scientific Director, Infection and Immune Reconstitution WC

How Do You Measure Success in ALL?: Assessment of MRD

Role of FISH in Hematological Cancers

Molecular Detection of BCR/ABL1 for the Diagnosis and Monitoring of CML

Test Name Results Units Bio. Ref. Interval. Positive

Whole Genome and Transcriptome Analysis of Anaplastic Meningioma. Patrick Tarpey Cancer Genome Project Wellcome Trust Sanger Institute

Acute leukemia and myelodysplastic syndromes

Acute Lymphoblastic and Myeloid Leukemia

Test Name Results Units Bio. Ref. Interval. Positive

Lymphoblastic Leukemia / Lymphoma

Mixed Phenotype Acute Leukemias

NUP214-ABL1 Fusion: A Novel Discovery in Acute Myelomonocytic Leukemia

MP BCR-ABL1 Testing in Chronic Myelogenous Leukemia and Acute Lymphoblastic Leukemia

NGS in tissue and liquid biopsy

Johann Hitzler, MD, FRCPC, FAAP Jacqueline Halton, MD, FRCPC Jason D. Pole, PhD

Blastic Plasmacytoid Dendritic Cell Neoplasm with DNMT3A and TET2 mutations (SH )

First relapsed childhood ALL Role of chemotherapy

2011: ALL Pre-HCT. Subsequent Transplant

Supplementary Figures and Tables

NEXT GENERATION SEQUENCING. R. Piazza (MD, PhD) Dept. of Medicine and Surgery, University of Milano-Bicocca

ONE STEP MULTIPLEX RT-PCR FOR BCRlABL GENE IN MALAY PATIENTS DIAGNOSED AS LEUKAEMIA

Corporate Medical Policy. Policy Effective February 23, 2018

CAALL-F01 OVERVIEW, OUTLINES COMITE LEUCEMIES 19 NOVEMBRE 2015

SWOG ONCOLOGY RESEARCH PROFESSIONAL (ORP) MANUAL LEUKEMIA FORMS CHAPTER 16A REVISED: DECEMBER 2017

ADVANCES IN CHILDHOOD ACUTE LEUKEMIAS : GENERAL OVERVIEW

Genomic Medicine: What every pathologist needs to know

JAK2 V617F analysis. Indication: monitoring of therapy

HD-SNP Microarray Analysis of the Study 9 High Risk ALL Patients -

SALSA MLPA probemix P383-A1 T-ALL Lot A

Addressing the challenges of genomic characterization of hematologic malignancies using microarrays

Acute myeloid leukemia. M. Kaźmierczak 2016

Summary. Olga Zając, Katarzyna Derwich, Katarzyna Stefankiewicz, Jacek Wachowiak. Rep Pract Oncol Radiother, 2007; 12(5):

p.r623c p.p976l p.d2847fs p.t2671 p.d2847fs p.r2922w p.r2370h p.c1201y p.a868v p.s952* RING_C BP PHD Cbp HAT_KAT11

Supplementary Figure 1. Successful excision of genes from WBM lysates and

MPL W515L K mutation

Case 1. Sa A.Wang, MD UT MD Anderson Cancer Center Houston, TX

A novel isothermal amplification approach for rapid identification of BCR-ABL fusion genes at onset:

Laboratory Service Report

Acute myeloid leukemia: prognosis and treatment. Dimitri A. Breems, MD, PhD Internist-Hematoloog Ziekenhuis Netwerk Antwerpen Campus Stuivenberg

sequences of a styx mutant reveals a T to A transversion in the donor splice site of intron 5

Controversies in Hematology: Case-Based Discussion. Acute leukemia in Adolescents and Young adults October 2018, Chiang Mai Thailand

Concomitant WT1 mutations predicted poor prognosis in CEBPA double-mutated acute myeloid leukemia

Correlation of Sex and Remission of Acute Lymphoblastic Leukemia-L1 (ALL-L1) in Children

Myelodysplasia/Myeloproliferative Neoplasms (MDS/MPN) Post-HCT Data

Supplemental Information For: The genetics of splicing in neuroblastoma

Nature Medicine: doi: /nm.4439

FAST FACTS Eligibility Reviewed and Verified By MD/DO/RN/LPN/CRA Date MD/DO/RN/LPN/CRA Date Consent Version Dated

Next Generation Sequencing in Haematological Malignancy: A European Perspective. Wolfgang Kern, Munich Leukemia Laboratory

The Human Major Histocompatibility Complex

Please Silence Your Cell Phones. Thank You

Molecular Hematopathology Leukemias I. January 14, 2005

Pearson r = P (one-tailed) = n = 9

SALSA MLPA probemix P241-D2 MODY mix 1 Lot D As compared to version D1 (lot D1-0911), one reference probe has been replaced.

Nature Genetics: doi: /ng Supplementary Figure 1. Details of sequencing analysis.

BWA alignment to reference transcriptome and genome. Convert transcriptome mappings back to genome space

Case Report RCSD1-ABL1 Translocation Associated with IKZF1 Gene Deletion in B-Cell Acute Lymphoblastic Leukemia

Analysis of Massively Parallel Sequencing Data Application of Illumina Sequencing to the Genetics of Human Cancers

Application of Whole Genome Microarrays in Cancer: You should be doing this test!!

Nature Genetics: doi: /ng Supplementary Figure 1. HOX fusions enhance self-renewal capacity.

Handout for lecture on lymphoblastic neoplasms presented by Rob McKenna

Reporting TP53 gene analysis results in CLL

Clinical Guidelines for Lymphoid Diseases Acute Lymphoblastic Leukaemia (ALL)

Aberrant Expression of CD7 in Myeloblasts Is Highly Associated With De Novo Acute Myeloid Leukemias With FLT3/ITD Mutation

Supplementary Appendix

Nature Genetics: doi: /ng Supplementary Figure 1. Clinical timeline for the discovery WES cases.

KYMRIAH (tisagenlecleucel)

SALSA MLPA probemix P241-D2 MODY mix 1 Lot D2-0716, D As compared to version D1 (lot D1-0911), one reference probe has been replaced.

Oncogenes. Dr. S Hosseini-Asl

Supplementary Figure 1. Generation of knockin mice expressing L-selectinN138G. (a) Schematics of the Sellg allele (top), the targeting vector, the

Nature Genetics: doi: /ng Supplementary Figure 1. Somatic coding mutations identified by WES/WGS for 83 ATL cases.

Diagnostic challenge: Acute leukemia with biphenotypic blasts and BCR-ABL1 translocation

Myeloproliferative Disorders - D Savage - 9 Jan 2002

Supplementary Figure 1. Gating strategy for leukemic blasts and normal developing B

MANAGEMENT OF ACUTE LYMPHOBLASTIC LEUKEMIA. BY Dr SUBHASHINI 1 st yr PG DEPARTMENT OF PEDIATRICS

Supplementary appendix

Case #1. 65 yo man with no prior history presented with leukocytosis and circulating blasts: Bone marrow biopsy was performed

All patients with FLT3 mutant AML should receive midostaurin-based induction therapy. Not so fast!

Philadelphia chromosome-positive acute lymphoblastic leukemia in childhood

Celebrating 20 years of the Database Joint UKCCG and CHO Annual Conference. 22 nd -23 rd March 2012 Newcastle upon Tyne

Personalized Therapy for Acute Myeloid Leukemia. Patrick Stiff MD Loyola University Medical Center

P323-B1 CDK4-HMGA2-MDM2

N Engl J Med Volume 373(12): September 17, 2015

The autoimmune disease-associated PTPN22 variant promotes calpain-mediated Lyp/Pep

MS.4/ Acute Leukemia: AML. Abdallah Al Abbadi.MD.FRCP.FRCPath Feras Fararjeh MD

Transcription:

Supplementary Figure 1 TCF3 breakpoints of TCF3-PBX1 (patients 1a 5a) and TCF3-HLF (patients 6a 9a and11a) translocations. The CpG motifs closest to the breakpoints are highlighted in red boxes and the closest cac motifs in blue.

Supplementary Figure 2 The TCF3-HLF translocation is only detected in the lymphoid progenitor gate. Experiments, combining sorting of specific cell populations using flow cytometry (a) and subsequent detection of the fusion gene in sorted cells employing FISH probes or real-time PCR (b), were performed using bone marrow from the TCF3-HLF-positive patients 15,

16, and 17 of the validation cohort. Bone marrow cells were sorted into HSC (CD34 + CD38 - CD90 + CD45RA - ) and MLP (CD34 + CD38 - CD90 neg-low CD45RA + ) populations as previously described (Doulatov et al. Nature Immunology 11, 585-593 (2010)). Leukemic cells (CD34 - CD38 + CD19 + CD10 + ) were sorted according to standard diagnostic markers. Sorted cell populations were analyzed by FISH and real-time PCR as described in the Online Methods section. All sorted leukemia cells carried the translocation (FISH signals 2R, 1G, 1B, 2B/G fusion) while none of the scored HSC or MLP (2R, 2G, 2B) populations were positive. The TCF3-HLF fusion was detected only in blast populations and not in myeloid cells indicating that the TCF3-HLF-positive leukemia originated in committed lymphoid cells. For patient 15, the results were confirmed using a TCF3-HLF-fusion-specific quantitative real-time PCR on a fraction of sorted cells as described in the supplementary online methods section. Leukemic cells were 100% positive for the TCF3-HLF-fusion, whereas the MLP and the HSC fractions were negative. As a positive control the reference gene -globin was employed. The TCF3-HLF fusion was detected by real-time PCR only in the blast population, but not in myeloid cells, indicating that the TCF3-HLF-positive leukemia may have originated from committed lymphoid cells.

Supplementary Figure 3 BTG1 deletions. (a) Whole genome sequencing (WGS) read ratio plot showing a BTG1 deletion identified in the diagnostic sample of patient 8. This BTG1 deletion was present in a subclonal population of ~14% of leukemia cells. (b) WGS read ratio plot showing a BTG1 deletion identified in the diagnostic sample of patient 9. (c) Exome sequencing read ratio plot for a xenograft (7b_X) derived from the remission sample of patient 7 showing a BTG1 deletion. (d) The BTG1 deletion in xenograft 7b_X ia also visible in the SNP allele frequency data from exome sequencing, inspecting the SNP rs709222 in BTG1 exon 2.

a chromosome 1p35.1 223 kb deletion in 6a chr1: 32.50 32.55 32.60 32.65 32.70 Mbp [hg19] KHDRBS1 TMEM39B KPNA6 DCDC2B LCK TXLNA EIF3I CCDC28B FAM167B IQCC b Fusion border RNA-seq read coverage (RPM) 1.2 0 KHDRBS1 c 0.8 0 LCK isoform 2 FAM167B LCK isoform 1 d chr1:32479954-32479978(+)...cacgccatgcagctgctgacggcag chr1:32739926-32739950(+) GGACCATGGGCTGTGGCTGCAGCTC... fused exons e GCTCGACCCGTCCTTCACTCACGCCATGCAGCTGCTGACGGCAG GTCCTTCACTCACGCCATGCAGCTGCTGACGGCAG CTTCACTCACGCCATGCAGCTGCTGACGGCAG ACTCACGCCATGCAGCTGCTGACGGCAG GGACCAT (1x) GGACCATGGGCTGTGG (1x) GGACCATGGGCTGTGGCTG (1x) GGACCATGGGCTGTGGCTGCAGC (1x) RNA-seq reads on fusion TGCAGCTGCTGACGGCAG CTGCTGACGGCAG GCTGACGGCAG GGACCATGGGCTGTGGCTGCAGCTCACACCCGG (5x) GGACCATGGGCTGTGGCTGCAGCTCACACCCGGAAGAT (3x) GGACCATGGGCTGTGGCTGCAGCTCACACCCGGAAGATGA (1x) CTGACGGCAG GGACCATGGGCTGTGGCTGCAGCTCACACCCGGAAGATGAC (1x) KHDRBS1 exon 1... D P S F T H A M Q L L T A LCK exon 2 (in-frame) fusion protein G T M G C G C S S H P E D D... Supplementary Figure 4 KHDRBS1-LCK fusion gene in patient 6a. (a) A heterozygous deletion encompassing 223 kb of genomic DNA on chromosome 1 fused intron 1 of KHDRBS1 to the promoter region of the LCK proto-oncogene. (b) Transcriptome sequencing showed a decrease of KHDRBS1 expression after exon 1. (c) LCK expression was induced starting from exon 2. (d) Analysis of chimeric fusion reads showed the fusion of KHDRBS1 exon 1 with LCK exon 2, confirmed by 14 chimeric reads spanning the fusion border. (e) Amino acid sequence of the in-frame fusion protein encoded by the KHDRBS1-LCK fusion transcript.

Supplementary Figure 5 Recurrent fusion of the genes KHDRBS1 and LCK in ALL. RNA was isolated from samples of a cohort of 74 unselected pediatric ALL and transcribed into cdna. Nested PCR was performed using specific primers for the KHDRBS1-LCK fusion transcript detected in patient 6a. (a) The gel electrophoresis presents the expected specific PCR product of 226 bp in the sample 6a and three patient samples of the validation cohort as well as controls. The negative control is representative of the 71 patient samples of this validation cohort tested negative. (b) The chromatograms display the sequencing result for patient 6a (upper panel) and a patient of the validation cohort (patient D3, lower panel) as an example. The fusion of KHDRBS1 exon 1 to LCK exon 2 was confirmed by Sanger sequencing in all positive cases (3/74).

Supplementary Figure 6 Pattern of recurrent genomic lesions and model of leukemogenesis in TCF3-HLF positive leukemia. Left panel: Recurrent genetic lesions identified in TCF3-HLF-positive ALL affect transcription factors regulating lymphoid differentiation, the Ras pathway and cell cycle regulation. Besides the 12 cases analyzed in the present study, a single case reported in a recent study is included (Ma et al. Nature Communications 6, 6604 (2015), DOI: 10.1038/ncomms7604) (x indicates a KRAS mutation that was detected only in a patient derived xenograft sample, not in the primary tissue. Note that the cases 16 and 17 have not been analyzed by next generation sequencing and besides PAX5 deletion, NRAS and KRAS hotspot mutations other genetic alterations possibly present are not yet analyzed). Right panel: The TCF3-HLF gene fusion probably occurs in an early B cell progenitor. B cell differentiation is blocked by mutations in transcription factors playing key roles in lymphoid development. Secondary mutations involving the Ras pathway and cell cycle regulators lead to leukemia development.

Supplementary Figure 7 Engraftment of TCF3-PBX1 and TCF3-HLF positive ALL samples. ALL cells were recovered from cryopreserved samples and transplanted into NSG mice. The percentage of mcd45 - hcd19 + hcd45 + leukemic cells was monitored by flow cytometry every 4 weeks. Upper panels: Primary and secondary transplantation of diagnostic TCF3-PBX1-positive samples. Middle panels: Primary and secondary transplantation of diagnostic TCF3-HLF-positive samples. Lower panels: Primary and secondary transplantation of MRD and relapse TCF3-HLF-positive samples. MRD samples were successfully amplified only for the TCF3-HLF-positive subtype. Patient samples were transplanted intrafemorally (primary transplantation). Xenograft-samples were transplanted intravenously (secondary transplantation). Mean and SD of 2-3 mice is shown when available. The number of living cells transplanted is indicated in the legend.

Supplementary Figure 8 Expression of PAX5 in primary and patient derived xenografts (PDX) of TCF3-PBX1 and TCF3-HLF positive ALL samples. The significant differential expression of PAX5 between TCF3-PBX1-positive and TCF3-HLF-positive primary ALL is preserved in patient derived xenograft samples.

Supplementary Figure 9 Cell survival and cell cycle analysis of TCF3-PBX1 and TCF3-HLF positive ALL cells in coculture on hmscs. Upper panel: ALL cell viability comparing co-cultures to ALL cell suspension cultures after 72 h by flow cytometry using 7-AAD staining. The percentage of living cells normalized to the input cells seeded on day 0 is shown (mean with SD of duplicates). Lower panel: Percentage of cells of the population in the G 0 G 1, S and G 2 M phases of the cell cycle. Flow cytometric analysis was performed at day 1 of co-culture with hmsc after 20 h of EdU incubation. Mean and SD of at least 2 independent experiments performed in triplicates are shown.

Supplementary Figure 10 Drug profiling of TCF3-PBX1 and TCF3-HLF ALL xenografts derived from diagnostic, MRD and relapse samples. Unsupervised clustering of xenograft samples according to their response (log IC 50 ) to 98 compounds in co-culture on hmsc after 72 h. Xenografts derived from diagnostic a, MRD b and relapse c samples are included. Fitted values are provided in the supplementary section (absolute IC 50 ). Compounds are arranged based on the family of the targets. Numbers identify the compounds shown in Figure 5c.

Supplementary Figure 11 In vitro dose response curves to ABT-199 (venetoclax) comparing xenografts derived from TCF3-HLF-positive ALL patients at the time point of diagnosis 'a', MRD 'b' and relapse 'c'. ALL co-cultures were treated with ABT-199 for 72 hours, the responses were normalized against DMSO-treated controls..

Supplementary Figure 12 Drug activity profiling of two additional TCF3-HLF translocated leukemia cases of the validation cohort (15, 17) reveals similar patterns of drug sensitivities and high responsiveness to ABT-199 (venetoclax) compared to the TCF3-HLF positive patients of the discovery cohort. Selection of drugs based on differences in sensitivity or resistance in TCF3-PBX1-positive compared to TCF3-HLF-positive patients (Figure 5c). Similar patterns of drug sensitivity were observed testing patient derived primary (patient 15, 17) and xenograft cells (other TCF3-PBX1- and TCF3-HLF-positive cases shown). Drugs active across both patient cohorts include doxorubicin, idarubicin, mitoxantrone, bortezomib, and ABT-199 (venetoclax).

Supplementary Figure 13 In vitro dose response co-titration curves to ABT-199/dexamethasone (top) and ABT-199/vincristine (bottom) for patient derived xenografts from TCF3-HLF positive (6a 11a) ALL patients. Xenograft cells were seeded on a layer of stroma cell and treated with the indicated combinations and concentrations of ABT-199, vincristine and dexamethasone, respectively, for 72 h. Drug concentrations used in these assays are provided in Supplementary Table 26. The response was normalized against DMSO-treated controls. The co-titration curves indicate that ABT-199 combined with either vincristine or dexamethasone likely exhibits synergistic activity in the patients marked with (*); combination indices (see Table) were calculated based on the Chou-Talaly method (Chou and Talaly JBC 252: 6438-6442, (1977)).

Supplementary Table 1. Characteristics of patients with TCF3-PBX1 and TCF3-HLF fusion-positive pediatric acute lymphoblastic leukemia included in initial sequencing analyses. TCF3-PBX1 TCF3-HLF Patient 1 2 3 4 5 6 7 8 9 11 Sex male female male female female male female male male male Age at diagnosis (years) 15.5 14.5 10.8 1.8 15.5 16.5 14.5 14.1 13.8 15.5 WBC a /!l at diagnosis 26,800 90,600 10,370 102,000 15,520 14,300 2,200 6,300 5,800 20,300 CNS status b CNS1 CNS1 CNS1 CNS2 CNS1 CNS1 CNS1 CNS1 CNS1 CNS1 Prednisone response c good poor poor good poor good good poor good good Morphological remission in bone marrow after induction treatment d M1 M1 M1! M1! M1! M1! M1! M1! M1 M3 MRD after induction negative 10-4 treatment e Treatment outcome f CCR CCR after HSCT negative negative negative 10-3 10-3 10-1 10-3 10-2 CCR CCR CCR TRM TRM relapse and death after HSCT relapse and death relapse and death after HSCT Leukemic specimen g used for sequencing (% blasts) BM (96) PB (92) BM (98) BM (90) BM (94) BM (98) BM (81) BM (61) BM (72) BM (88) Normal reference tissue BM BM BM BM BM BM BM BM BM BM Treatment protocol h BFM BFM BFM! BFM! BFM! BFM BFM BFM FRALLE BFM a white blood cell count; b central nervous system (CNS) status: CNS1 (lumbar puncture nontraumatic, no leukemic blasts in the cerebrospinal fluid (CSF) after cytocentrifugation); CNS2 (puncture nontraumatic, "5 leukocytes/!l CSF with identifiable blasts; the prognostic impact of CNS2 status is debated); CNS3 (puncture nontraumatic, >5 leukocytes/!l CSF with identifiable blasts); TLP+ (traumatic lumbar puncture with identifiable leukemic blasts); a TLP with no identifiable blasts is not an adverse factor. For cytomorphological examination, CSF samples should be analyzed after cytospin preparation, a method through which cellular components within the CSF are concentrated by centrifugation; c assessed after 7 days induction with daily prednisone and a single intrathecal dose of methotrexate on treatment day 1, good: < 1000/!l blood blasts, poor: # 1000/!l blood blasts; d < 5% blasts (M1), # 5% blasts (M2 or M3); e assessed by DNA-PCR-based monitoring of leukemic immunoglobuline and/or T-cell receptor rearrangements, markers required to have a sensitivity of at least 10-4 ; f CCR: continuous complete remission, HSCT: hematopoietic stem cell transplantation, TRM: treatment-related mortality; g BM: bone marrow, PB: peripheral blood; h BFM: Berlin-Frankfurt-Münster study group.!!!

Supplementary Table 2. Characteristics of TCF3-HLF-positive pediatric acute lymphoblastic leukemia patients included in validation analysis. Patient 10 12 13 14 15 16 17 Sex female female male male male female female Age at diagnosis (years) 12.7 18.2 14.7 3.4 13.6 14.3 14 WBC a /µl at diagnosis 5,000 5,200 unknown 4,240 3,470 6,150 4,450 CNS status b CNS1 CNS1 CNS1 CNS1 CNS1 CNS1 CNS1 Prednisone response c not applicable not applicable good good good good not applicable Morphological remission in bone marrow after induction M1 M1 M1 M3 M1 M2 no treatment d 6x10 MRD after induction treatment e 10-1 unknown negative 3.5x10-1 1x10-4 10-1 unknown Treatment outcome f relapse and death after SCT relapse and death after SCT relapse and death relapse and death after SCT -1 remission after SCT relapse after HSCT primary refractory Leukemic specimen g used for sequencing (% blasts) BM (53) BM (unknown) PB (79) BM (80) BM (93.5) not applies not applies Normal reference tissue BM BM BM not available not available not applies not applies Treatment protocol h UKALL UKALL FRALLE BFM BFM BFM AALL1131 PAX5 deletion/mutation no no yes yes yes unknown unknown BTG1 deletion/mutation uncertain yes no no no unknown unknown VPREB1 deletion no yes yes no no unknown unknown TCF3 mutation - - S467G - - unknown unknown RAS pathway mutation i - SPHK1 P16H - PTPN11 T73I PTPN11 Q86R unknown unknown

a white blood cell count; b central nervous system (CNS) status: CNS1 (lumbar puncture nontraumatic, no leukemic blasts in the cerebrospinal fluid (CSF) after cytocentrifugation); CNS2 (puncture nontraumatic, 5 leukocytes/µl CSF with identifiable blasts; the prognostic impact of CNS2 status is debated); CNS3 (puncture nontraumatic, >5 leukocytes/µl CSF with identifiable blasts); TLP+ (traumatic lumbar puncture with identifiable leukemic blasts); a TLP with no identifiable blasts is not an adverse factor. For cytomorphological examination, CSF samples should be analyzed after cytospin preparation, a method through which cellular components within the CSF are concentrated by centrifugation; c assessed after 7 days induction with daily prednisone and a single intrathecal dose of methotrexate on treatment day 1, good: < 1000/µl blood blasts, poor: 1000/µl blood blasts; d < 5% blasts (M1), 5% blasts (M2 or M3); e assessed by DNA-PCR-based monitoring of leukemic immunoglobuline and/or T-cell receptor rearrangements, markers required to have a sensitivity of at least 10-4 ; f CCR: continuous complete remission, HSCT: hematopoietic stem cell transplantation, TRM: treatment-related mortality; g BM: bone marrow, PB: peripheral blood; h BFM: Berlin-Frankfurt-Münster study group.

Supplementary Table 3. Characteristics of patients with TCF3-PBX1 fusion-positive pediatric acute lymphoblastic leukemia included in validation analyses a. TCF3-PBX1 (n = 24) n (%) Sex male 11 (45.8) female 13 (54.2) Age at diagnosis (years) < 1-1 - 9 14 (58.3) # 10 10 (41.7) WBC b /!l at diagnosis < 10,000 3 (12.5) 10,000 49,999 15 (62.5) 50,000 99,999 3 (12.5) #100,000 3 (12.5) Prednisone response c good 20 (83.3) poor 4 (16.7) MRD d TP 1 + 2 negative 9 (37.5) other 10 (41.7) TP 2 # 10-3 4 (16.7) unknown 1 (4.2) EBF1 deletion e negative 16 (66.7) positive 3 (12.5) unknown 5 (20.8) CDKN2A/B deletion e negative 16 (66.7) positive 3 (12.5) unknown 5 (20.8) PAX5 deletion e negative 18 (75.0) positive 1 (4.2) unknown 5 (20.8) ETV6 deletion e negative 18 (75.0) positive 1 (4.2) unknown 5 (20.8) RAS pathway mutation f negative 23 (95.8) positive 1 (4.2) Relapse no 22 (91.7) yes 2 (8.3)

a 24 patients included in validation of RAS pathway mutation screening, 19 patients included in validation of deletions associated with B-cell differentiation (e.g. PAX5) by MLPA; b white blood cell count; c assessed after 7 days induction with daily prednisone and a single intrathecal dose of methotrexate on treatment day 1, good: < 1000/!l blood blasts, poor: # 1000/!l blood blasts; d MRD: minimal residual disease as assessed by DNA-PCR-based monitoring of leukemic immunoglobuline and/or T-cell receptor rearrangements, markers required to have a sensitivity of at least 10-4 ; TP: time point, TP1: MRD after induction at treatment day 33, TP2: MRD at treatment day 78, MRD #10-3 qualifies for the high-risk group; e five out of 19 patients affected, one patient harbored several deletions (CDKN2A and B, PAX5, ETV6, BTG1), one patient demonstrated co-occurrence of EBF1 and CDKN2A and B, the remaining three patients had single occurrences of EBF1 (n=2) or CDKN2A and B; f RAS pathway screening by Sanger sequencing included: KRAS exon 1, NRAS exon 1 and exon 2, FLT3 exon 14 and 20, PTPN11 exons 3 and 13, one patient found positive for an NRAS exon 2, codon 61 (CAA>CGA, Q61R) activating mutation.!!!

Supplementary Table 4. Genomic breakpoint coordinates, fused exons and nucleotides at the junctions of TCF3-PBX1 and TCF3-HLF gene fusions detected in pediatric ALL patient samples. Coordinates of breakpoints were obtained from Sanger sequencing of PCR products from genomic DNA or cdna. Ins=Insertion; Mhom=microhomology; TpIns=Templated insertion [*:Chr17; $:ChrX].

Supplementary Table 8. Characteristics of pediatric acute lymphoblastic leukemia patients from trial AIEOP-BFM ALL 2000 included in mutation screening of TCF3 exon 19. AIEOP-BFM ALL 2000 cohort n = 1033 n (%) Sex male 593 (57.4) female 440 (42.6) Age at diagnosis (years) < 1 1 (0.1) 1-9 749 (72.5)! 10 283 (27.4) WBC a /"l at diagnosis < 10,000 405 (39.2) 10,000 49,999 365 (35.3) 50,000 99,999 128 (12.4)!100,000 135 (13.1) Prednisone response b good 899 (87.0) poor 126 (12.2) unknown 8 (0.8) MRD c TP 1 + 2 negative 359 (34.8) other 433 (41.9) TP 2! 10-3 65 (6.3) unknown 176 (17.0) Immunophenotype B-cell 840 (81.3) T-cell 181 (17.5) unknown/other 12 (1.2) BCR/ABL1 negative 1014 (98.2) positive 19 (1.8). MLL/AF4 negative 1029 (99.6) positive 4 (0.4) ETV6/RUNX1 negative 754 (73.0) positive 208 (20.1) unknown 71 (6.9) a white blood cell count; b assessed after 7 days induction with daily prednisone and a single intrathecal dose of methotrexate on treatment day 1, good: < 1000/"l blood blasts, poor:! 1000/"l blood blasts; c MRD: minimal residual disease as assessed by DNA-PCR-based monitoring of leukemic immunoglobuline and/or T-cell receptor rearrangements, markers required to have a sensitivity of at least 10-4 ; TP: time point, TP1: MRD after induction at treatment day 33, TP2: MRD at treatment day 78, MRD!10-3 qualifies for the high-risk group.!!