-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.

567 RNA Splicing Modulation Impairs Acute Myeloid Leukemia Stem Cell Maintenance

Acute Myeloid Leukemia: Novel Therapy, excluding Transplantation
Program: Oral and Poster Abstracts
Type: Oral
Session: 616. Acute Myeloid Leukemia: Novel Therapy, excluding Transplantation: Novel Targeting Approaches
Monday, December 7, 2015: 11:00 AM
W109, Level 1 (Orange County Convention Center)

Leslie A Crews, PhD1,2*, Larissa Balaian, Ph.D.3, Heather Leu, BS2*, Nathaniel Delos Santos2*, Angela C Court2*, Anil Sadarangani, PhD4*, Maria A. Zipeto, PhD2*, James J La Clair5*, Reymundo Villa5*, Sheldon Morris, MD6*, Rainer F. Storb, MD7, Anna Kulidjian, MD8*, Edward D. Ball, MD9, Michael D Burkart, PhD5* and Catriona Jamieson, MD, PhD1,2,3

1Sanford Consortium for Regenerative Medicine, La Jolla, CA
2Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA
3Moores Cancer Center, University of California, San Diego, La Jolla, CA
4Division of Regenerative Medicine, University of California San Diego, La Jolla, CA
5Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA
6Department of Medicine, University of California, San Diego, La Jolla, CA
7Fred Hutchinson Cancer Research Center, Seattle, WA
8Department of Orthopedic Surgery, University of California San Diego, La Jolla, CA
9Division of Bone Marrow Transplantation, Department of Medicine, University of California, San Diego, La Jolla, CA

Introduction

Disease relapse is the leading cause of death in secondary AML (sAML), which evolves from antecedent hematologic disorders like myelodysplastic syndrome (MDS) or myeloproliferative neoplasms (MPNs) or following exposure to chemotherapy. Persistence of therapy-resistant leukemia stem cells (LSC) harboring enhanced survival and self-renewal capacity has been linked to high relapse rates in sAML. Previously, we showed that missplicing of a stem cell regulatory gene, GSK3b, and splice isoform switching favoring pro-survival BCL2 family isoform expression promoted generation of therapy-resistant LSC (Abrahamsson et al PNAS 2009; Goff et al Cell Stem Cell 2013). However, whether aberrant pre-mRNA splicing promotes sAML LSC generation, in the absence of mutation, and if pharmacological splicing modulation impairs LSC maintenance, in a mutation-independent manner, has not been elucidated.

Methods and Results

Comparative RNA-sequencing and gene set enrichment analyses revealed significant alterations in splicing factor gene expression in purified progenitors from untreated sAML compared with normal samples. In addition, using an isoform-specific alignment algorithm, we established a sAML LSC splice isoform expression signature that identified increased expression of select transcripts, e.g. CD82 and PTK2B. Thus, we investigated the LSC inhibitory efficacy of a stable, potent splicing modulatory agent, 17S-FD-895, in humanized AML LSC stromal co-culture and primagraft assays. Notably, there was a dose-dependent reduction in AML LSC (n=4) survival and self-renewal after in vitro 17S-FD-895 treatment, with a favorable therapeutic index compared to normal controls (n=3, p<0.01). Splicing reporter activity and PCR analyses revealed rapid and potent 17S-FD-895-induced alterations in splicing, promoting pro-apoptotic isoform expression and intron inclusion in the stem cell regulatory gene MCL1. Also, 17S-FD-895 restored normal expression patterns of PTK2B, and MCL1-L/S and BLC2-L/S expression ratios.

Flow cytometric analyses in AML LSC primagraft models treated with 17S-FD-895 (5-10 mg/kg delivered intravenously in 3 doses over 2 weeks) revealed a decrease in human stem (CD45+CD34+CD38- Lin-, 68% reduction in the spleens of the 10 mg/kg group versus vehicle controls, n=5 mice per group, p<0.05) and progenitor (CD45+CD34+CD38+ Lin-, 80% reduction to nearly zero in the spleens of the 10 mg/kg group versus vehicle controls, p=0.08) cell frequencies. Furthermore, MCL1-L/S and BCL2-L/S expression ratios were significantly reduced in LSC-enriched fractions from 17S-FD-895-treated mice compared to vehicle controls. Consistent with a reduction in functional LSC burden after 17S-FD-895 treatment, subsequent serial transplantation studies showed a 47-65% reduction in leukemic burden in the hematopoietic tissues of recipients of CD34+ cells from mice in the 10 mg/kg treatment group versus vehicle controls (n=5 mice per group, p<0.05).

Conclusions

Here we demonstrate that a potent and stable splicing modulatory agent, 17S-FD-895, normalized sAML-specific splice isoform expression patterns as well as MCL1-L/S and BLC2-L/S ratios. Moreover, pharmacologic splicing modulation reduced AML LSC survival and self-renewal in a dose-dependent manner in both in vitro and in vivo models with a favorable therapeutic index. Further evaluation of this compound as a splicing-targeted single agent or combined with standard of care therapy may reduce or eradicate LSC burden in therapy-resistant sAML. In addition, LSC-specific splice isoforms may represent important biomarkers that could be developed as companion diagnostics for splicing-targeted therapies in sAML and other recalcitrant malignancies.

Disclosures: Ball: Regimmune: Research Funding . Jamieson: GlaxoSmithKline: Research Funding ; Johnson & Johnson: Research Funding ; CTI Biopharma: Research Funding .

*signifies non-member of ASH