-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.

3627 RNA Sequencing Reveals Novel and Rare Fusion Transcripts in Acute Myeloid Leukemia

Chromosomal Rearrangements and DNA Repair
Program: Oral and Poster Abstracts
Session: 601. Chromosomal Rearrangements and DNA Repair: Poster III
Monday, December 7, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Antonella Padella1*, Giorgia Simonetti, PhD1*, Giulia Paciello2*, Anna Ferrari, PhD1*, Elisa Zago, PhD3*, Carmen Baldazzi, PhD1*, Viviana Guadagnuolo, PhD1*, Cristina Papayannidis, MD, PhD4, Valentina Robustelli5*, Enrica Imbrogno5*, Nicoletta Testoni1*, Gerardo Musuraca6*, Simona Soverini7, Massimo Delledonne, PhD3*, Ilaria Iacobucci, PhD8*, Clelia Tiziana Storlazzi, PhD9*, Elisa Ficarra, PhD2* and Giovanni Martinelli8

1University of Bologna, Bologna, Italy
2Dept. of Control and Computer Engineering DAUIN, Politecnico di Torino, Turin, Italy
3Personal Genomics, Verona, Italy
4Bologna University School of Medicine, Bologna, Italy
5DIMES, Department of Hematology and Medical Sciences “L. and A. Serągnoli”, University of Bologna, Bologna, Italy
6Istituto Scientifico Romagnolo Per Lo Studio E La Cura Dei Tumori, Meldola (FC),, Meldola, Italy
7Institute of Hematology, S.Orsola-Malpighi University Hospital, Bologna, Italy
8"Serągnoli" Institute of Hematology, University of Bologna, Bologna, Italy
9Department of Biology, University of Bari, Bari, Italy

Acute Myeloid Leukemia (AML) is a highly heterogeneous disease and a complex network of events contribute to its pathogenesis. Chromosomal rearrangements and fusion genes have a crucial diagnostic, prognostic and therapeutic role in AML. A recent RNA sequencing (RNAseq) study on 179 AML revealed that fusion events occur in 45% of patients. However, the leukemogenic potential of these fusions and their prognostic role are still unknown.

To identify novel rare gene fusions having a causative role in leukemogenesis and to identify potential targets for personalized therapies, transcriptome profiling was performed on AML cases with rare and poorly described chromosomal translocations.

Bone marrow samples were collected from 5 AML patients (#59810, #20 and #84 at diagnosis and #21 and #32 at relapse). RNAseq was performed using the Illumina Hiseq2000 platform. The presence of gene fusions was assessed with deFuse and Chimerascan. Putative fusion genes were prioritized using Pegasus and Oncofuse, in order to select biologically relevant fusions. Chimeras not supported by split reads, occurring in reactive samples, involving not annotated or conjoined genes were removed. The remaining fusions were prioritized according to mapping of partner genes to chromosomes involved in the translocation or to Chimerascan and deFuse concordance.

The CBFβ-MYH11 chimera was identified in sample #84, carrying inv(16) aberration, thus confirming the reliability of our analysis.

Sample #59810 carried the fusion transcript ZEB2-BCL11B (Driver Score, DS=0.7), which is an in-frame fusion and a rare event in AML associated with t(2;14)(q21;q32). The breakpoint of the fusion mapped in exon 2 of ZEB2 (ENST00000558170) and exon 2 of BCL11B (ENST00000357195). Differently from previous data, this fusion transcript showed 3 splicing isoforms. Type 1 isoform is the full-length chimera and it retains all exons of both genes involved in the translocation. Type 2 isoform was characterized by the junction of exon 2 of ZEB2 and exon 3 of BCL11B. In type 3 isoform, exon 2 and 3 of BCL11B were removed, resulting in an mRNA composed by exon 2 of ZEB2 and exon 4 of BCL11B. Gene expression profiling showed an upregulation of ZEB2 and BCL11B transcripts in the patient’s blasts, compared to 53 AML samples with no chromosomal aberrations in the 14q32 region. The same samples showed the WT1-CNOT2 chimera, which is a novel out-of-frame fusion (DS= 0.008) related to t(11;12) translocation, identified by cytogenetic analysis.

Two new in-frame fusion genes were identified in sample #20: CPD-PXT1 (DS= 0.07), which appeared as the reciprocal fusion product of t(6;17) translocation, and SAV1-GYPB, which remained cryptic at cytogenetic analysis (DS= 0.8, alternative splicing events are being investigated). SAV1 was downregulated in sample #20 compared to our AML cohort, suggesting the putative loss of a tumour-suppressor gene.

Sample #21 carried a t(3;12) translocation and RNAseq identified a novel fusion event between chromosomes 19 and 7, involving the genes OAZ and MAFK (DS= 0.9). Finally, no chimeras were confirmed in sample #32 having a t(12;18) translocation.

Our data suggest that fusion events are frequent in AML and a number of them cannot be detected by current cytogenetic analyses. Gene fusions cooperate to AML pathogenesis and heterogeneity and we are further investigating the oncogenic potential of the identified translocations. Moreover, the results firmly indicate that different approaches, including G-banding, molecular biology, bioinformatics and statistics, need to be integrated in order to better understand AML pathogenesis and improve patients’ stratification, High-resolution sequencing analysis currently represent the most informative strategy to tailor personalized therapies.

Acknowledgements: ELN, AIL, AIRC, progetto Regione-Università 2010-12 (L. Bolondi), Fondazione del Monte di Bologna e Ravenna, FP7 NGS-PTL project.

Disclosures: Soverini: Novartis, Briston-Myers Squibb, ARIAD: Consultancy . Martinelli: BMS: Speakers Bureau ; MSD: Consultancy ; Roche: Consultancy ; ARIAD: Consultancy ; Novartis: Speakers Bureau ; Pfizer: Consultancy .

*signifies non-member of ASH