-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

693 Expression of an Oncogenic ERG isoform Characterizes a Distinct Subtype of B-Progenitor Acute Lymphoblastic Leukemia

Acute Lymphoblastic Leukemia: Biology, Cytogenetics and Molecular Markers in Diagnosis and Prognosis
Program: Oral and Poster Abstracts
Type: Oral
Session: 618. Acute Lymphoblastic Leukemia: Biology, Cytogenetics and Molecular Markers in Diagnosis and Prognosis: New Genomic Discoveries in Acute Lymphoblastic Leukemia
Monday, December 7, 2015: 3:15 PM
W331, Level 3 (Orange County Convention Center)

Jinghui Zhang, PhD1*, Kelly McCastlain2*, Chunxu Qu, PhD1*, Gang Wu, PhD1*, Michael Edmonson, PhD1*, Yongjin Li, PhD1*, Lei Wei, PhD2*, Debbie Payne-Turner, BS2*, Hiroki Yoshihara, MD PhD2*, Michelle L. Churchman, PhD2*, Esmé Waanders, PhD2,3, Panagiotis Ntziachristos, PhD4, Iannis Aifantis, PhD4,5, Kathryn G. Roberts2, Jing Ma, PhD2*, Guangchun Song, PhD2*, John Easton, PhD6*, Heather L. Mulder, BS6*, Xiang Chen, PhD1*, Michael Rusch, BA1*, Kristy Boggs, PhD6*, Bhavin Vadodaria, BS6*, James Dalton, BS2*, Marcus L Valentine, PhD7*, Li Ding, PhD8*, Charles Lu, PhD8*, Robert S. Fulton, PhD8*, Lucinda Fulton, PhD8*, Yashodan Tabib, PhD8*, Kerri Ochoa8*, Meenakshi Devidas, PhD9, Deqing Pei10*, Cheng Cheng, Ph.D10*, William E. Evans, PharmD11, Ching-Hon Pui, MD12, Sima Jeha, MD13, Richard C. Harvey, PhD14, I-Ming L Chen, DVM, MS14, Cheryl L. Willman, MD14, Guido Marcucci, MD15, Clara D. Bloomfield, MD16, Jessica Kohlschmidt, PhD, BS, MS16, Krzysztof Mrozek, MD, PhD16, Elisabeth Paietta, PhD17, Martin S. Tallman, MD18, Wendy Stock, MD19, Peter M. Voorhees, MD20, Janis Racevskis, MD21*, Jacob M. Rowe, MD22, Selina Luger, MD23, Steven M. Kornblau, MD24, Sheila A Shurtleff, PhD2*, Susana C. Raimondi, PhD2, Elaine R Mardis, PhD8*, Richard K Wilson, PhD25*, Stephen P Hunger, MD26, Mignon L. Loh, MD27, James R. Downing, MD2,28* and Charles G. Mullighan2

1Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN
2Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
3Department of Human Genetics, Radboud University Medical Centre and Radboud Center for Molecular Life Sciences, Nijmegen, Netherlands
4Department of Pathology, New York University, New York, NY
5Howard Hughes Medical Institute, New York, NY
6Pediatric Cancer Genome Project, St. Jude Children's Research Hospital, Memphis, TN
7Cytogenetics Core Facility, St. Jude Children's Research Hospital, Memphis, TN
8McDonnell Genome Institute, Washington University, St Louis, MO
9Department of Biostatistics, Colleges of Medicine, Public Health & Health Profession, University of Florida, Gainesville, FL
10Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
11Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
12Oncology, St. Jude Children's Research Hospital, Memphis, TN
13Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
14Department of Pathology, The Cancer Research and Treatment Center, University of New Mexico, Albuquerque, NM
15Gehr Leukemia Center, City of Hope, Duarte, CA
16Comprehensive Cancer Center, The Ohio State University, Columbus, OH
17Cancer Center, Montefiore Medical Center North Division, Bronx, NY
18Memorial Sloan-Kettering Cancer Center, New York, NY
19University of Chicago Medical Center, Chicago, IL
20Div. of Hematology/Oncology, University of North Carolina, Chapel Hill, NC
21Department of Medicine (Oncology), Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY
22Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
23Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
24Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX
25Washington University School of Medicine, Saint Louis, MO
26Department of Pediatrics, Children’s Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
27Department of Pediatrics, Benioff Children’s Hospital, University of California at San Francisco, San Francisco, CA
28Administration, St. Jude Children's Research Hospital, Memphis, TN

Introduction. Prior studies have described a subset of B-progenitor ALL cases with a distinct gene expression profile and/or deletions involving ERG (encoding the ETS family member v-ets avian erythroblastosis virus E26 oncogene), however the relationship of these alterations and their role in leukemogenesis are poorly understood. We performed integrated genomic and epigenetic analyses, biochemical studies and leukemogenesis assays to define the genetic basis of this form of ALL.

Methods. We studied 1674 childhood, adolescent and young adult B-progenitor ALL cases with microarray gene expression profiling and/or RNA-sequencing data to enable the identification of ERG ALL by unsupervised clustering and predictive analysis of microarrays. Detailed genomic analysis was performed for 144 ERG ALL cases, including whole genome (N=38), exome (n=46) and/or RNA-sequencing (n=57) cases, and single nucleotide polymorphism array analysis. Epigenetic profiling, including whole genome bisulfite sequencing, chromatin immunoprecipitation and sequencing for ERG and histone modifications and ATAC-sequencing were performed for a subset of 8 xenografted ERG tumors and reference cell lines. ERG transcript expression was measured by analysis of RNA-seq analysis and quantitative RT-PCR assays, and by interrogation of TCGA and PCGP RNA-seq data. The function of ERG isoforms was evaluated by EMSA and transcriptional reporter assays, immunofluoresence, colony forming assays and retroviral bone marrow transplant assays.

Results. One hundred and forty four cases (8.6%) of B-ALL cases exhibited a distinct gene expression profile and lacked known chromosomal rearrangements (ERG ALL). Such cases had favorable outcome. Eighty cases (55.6%) had focal deletions of ERG with no evidence of oncogenic or chimeric ERG fusions. The deletions were most commonly heterozygous and involving exons 3-7 (n=27) or 3-9 (n=22) of 10 coding exons, and less commonly involving exon 1, or a larger region of the gene. No ERG deletions were identified in non-ERG ALL. Two cases harbored missense mutations in the ETS domain. Analysis of whole genome and exome sequencing data of 71 cases identified a high frequency of alterations of lymphoid transcription factors (46.5%; IKZF1 36.7%, PAX5 11.3%); mutation of transcription factors otherwise uncommon in ALL (21%; MYC, MYCBP2, MGA, ZEB2, GATA3); activation of signaling pathways, most commonly NRAS or KRAS (35.2%); cell cycle regulation (22.5%); and epigenetic modifiers (56.3%), most commonly KMT2D, SETD2, ARID2 and NCOR1. Notably, the five year event-free survival of ERG ALL cases with IKZF1 alterations exceeded 85% in both St Jude and Children’s Oncology Group cohorts.

We observed striking transcriptional deregulation at the ERG locus. Most (51/56) ERG-deleted cases expressed an ERG isoform encoded by a novel exon in intron 6 that splices in frame to distal exons, resulting in expression of a truncated C-terminal ERG protein that lacks the pointed and central regulatory domains, but retains the ETS and transactivation domain (ERGalt). ERGalt was also present in most (36/44) cases lacking an ERG deletion, and was strongly associated with presence of ERGalt protein in leukemic cells. We also identified expression of an Antisense Long non-coding RNA associated with the ERG locus (ALE) in ERG ALL. ERGalt and ALE were absent, or uncommonly expressed at very low levels in non-ERG ALL. ERGalt was absent, and ALE rarely expressed in non-ALL PCGP and TCGA samples.

ERGalt and point mutant ERG were retained in the nucleus, bound DNA targets and acted as competitive inhibitors of wild type (WT) ERG in transcriptional reporter assays. Lineage-negative Arf-null bone marrow cells transduced with ERG WT induced an aggressive erythro-megakaryoblastic leukemia; in contrast ERGalt induced an immature lymphoid progenitor leukemia.

Conclusions. Genomic alterations drive aberrant transcription of ERG, resulting on expression of a truncated, C-terminal oncogenic ERG protein. This represents a novel mechanism of transcription factor deregulation in leukemia. As a subset of ERG ALL cases lack ERG deletion, and as IKZF1 alterations are not associated with inferior outcome in this form of ALL, diagnostic approaches must incorporate gene expression profiling in addition to identification of ERG and IKZF1 alterations to accurately identify this form of leukemia.

Disclosures: Evans: Prometheus Labs: Patents & Royalties: Royalties from licensing TPMT genotyping . Stock: Gilead: Membership on an entity’s Board of Directors or advisory committees . Voorhees: Oncopeptides: Consultancy ; Onyx Pharmaceuticals: Research Funding ; GSK: Consultancy ; Oncopeptides: Research Funding ; Janssen: Research Funding ; A Takeda Oncology Company: Consultancy , Research Funding ; Celgene: Consultancy ; Millennium Pharmaceuticals: Consultancy , Research Funding ; Acetylon Pharmaceuticals, Inc.: Research Funding ; Novartis: Consultancy ; Array BioPharma: Consultancy ; GSK: Research Funding ; Celgene: Research Funding . Hunger: Spectrum Pharmaceuticals: Consultancy ; Jazz Pharmaceuticals: Consultancy ; Sigma Tau: Consultancy ; Merck: Equity Ownership . Mullighan: Incyte: Consultancy ; Amgen: Honoraria .

*signifies non-member of ASH