-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.

1264 Imetelstat Inhibits Telomerase and Prevents Propagation of ADAR1-Activated Myeloproliferative Neoplasm and Leukemia Stem Cells

Program: Oral and Poster Abstracts
Session: 635. Myeloproliferative Syndromes: Basic Science: Poster I
Hematology Disease Topics & Pathways:
Therapies, Combinations, Clinically relevant
Saturday, December 5, 2020, 7:00 AM-3:30 PM

Wenxue Ma, MD, PhD1*, Larissa Balaian, PhD2*, Phoebe Mondala, BS3*, Yudou He, BS4*, Cayla Mason, BS3*, Jessica Pham, B.S.3,5*, Jeremy Lee, BS4*, Raymond Diep, BS3*, Sanja Coso, PhD6*, Kathleen Fisch, PhD7*, Adam Mark, MS8*, Sheldon Morris, MD, MPH2*, Qingfei Jiang, PhD3,9, Thomas Whisenant, PhD7*, Aleksandra Rizo, MD, PhD10, Fei Huang, PhD11*, Mary Donohoe, PhD6*, Ludmil Alexandrov, PhD4* and Catriona Jamieson, MD, PhD3,12,13,14,15

1Department of Medicine, Moores Cancer Center, and Sanford Stem Cell Clinical Center, University of California San Diego, La Jolla, CA
2Moores Cancer Center, University of California San Diego, La Jolla, CA
3Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA
4University of California San Diego, La Jolla, CA
5Division of Regenerative Medicine, University of California, San Diego, Tustin, CA
6Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, and Sanford Stem Cell Clinical Center, University of California San Diego, La Jolla, CA
7Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA
8Center for Computational Biology & Bioinformatics (CCBB), University of California San Diego, La Jolla
9Division of Regenerative Medicine, UCSD, San Diego, CA
10Geron Corporation, Parsippany, NJ
11Geron Corporation, Menlo Park, CA
12Sanford Stem Cell Clinical Center, University California, San Diego, La Jolla, CA
13Professor of Medicine, University California, San Diego, La Jolla, CA
14CIRM Alpha Stem Cell Clinic, University California, San Diego, La Jolla, CA
15Moores Cancer Center, University of California, San Diego, La Jolla, CA

BACKGROUND

Clonal stem cell derived myeloproliferative neoplasms (MPNs) have a propensity to evolve to acute myeloid leukemia (AML). Deregulation of the innate immune deaminase associated with RNA1 (ADAR1) has been linked to malignant progression and therapeutic resistance. Increased expression of the stem cell gene, human telomerase reverse transcriptase (hTERT), has also been linked with malignant transformation. However, the combinatorial role of ADAR1 and hTERT in the evolution of MPN stem cells to therapy resistant acute myeloid leukemia stem cells (LSCs) and the capacity of a telomerase inhibitor, imetelstat, to prevent survival and self-renewal of pre-LSC and LSC had not been established. Recent clinical trials show early signs of efficacy of imetelstat in treatment of myelofibrosis (MF). However, its role in selectively inhibiting pre-LSC transformation to self-renewing LSC has not been elucidated. Here we show that targeting telomerase activity prevents pre-LSC and LSC maintenance both in vitro and in vivo, suggesting telomerase inhibition as an effective strategy for preventing MPN progression.

METHODS

To quantify hTERT level and ADAR1 activity in the setting of normal HSPC and MPN stem cell evolution, whole genome sequencing (WGS) analysis was performed on 76 normal and MPN blood CD34+ cells and matching saliva samples. Results were compared with RNA-seq of 100 FACS purified young, aged, MPN and AML CD34+CD38- stem cells and CD34+CD38+ progenitor cells. Confocal fluorescence microscopic evaluation of stem cell ADAR1 and hTERT localization, telomere length by Flow-FISH and telomerase activity by TRAP assays, lentiviral ADAR1 overexpression and shRNA knockdown were performed. In vitro stromal co-cultures, and humanized immunocompromised mouse models were established to determine the impact of imetelstat (a oligonucleotide inhibitor of telomerase) on normal, MPN stem cell and LSC maintenance.

RESULTS

Combined hTERT overexpression, ADAR1 activation and a significant reduction in telomere length correlated with accelerated stem cell aging during MPN progression to AML. Increased ADAR1 mediated adenosine to inosine (A-to-I) transcript editing coincided with accelerated telomere shortening in high risk MPN stem cells. Moreover, lentiviral ADAR1 overexpression enhanced pre-LSC engraftment. Treatment with imetelstat reduced MPN stem cell and LSC propagation in stromal co-cultures as well as in humanized mouse models commensurate with reduced hTERT expression levels and telomerase activity and decreased ADAR1 editing activity. Specifically, stromal co-culture assays revealed that combined treatment with dasatinib at 1 nM, and imetelstat at 1 µM or 5 µM significantly inhibited survival and replating of blast crisis (BC) CML progenitors compared with aged bone marrow progenitors (p < 0.001, ANOVA). As a single agent, imetelstat (5 µM) inhibited survival and replating of pre-LSC derived from myelofibrosis compared with normal bone marrow progenitor samples (p < 0.001, ANOVA). In pre-LSC MPN NSG-SGM mouse models established from 4 different MF samples, a significant reduction in proliferation of human CD45+ cells (p < 0.01, t test) was observed in bone marrow and spleen, when compared with vehicle control. Treatment of humanized LSC mouse models, established with 5 different BC CML, with 30 mg/kg of imetelstat, 3 times a week for 4 weeks resulted in a significant reduction in proliferation of malignant progenitors and human CD45+ cells (p < 0.001, ANOVA). As measured by a Flow-FISH assay, abnormal telomere length was reversed by imetelstat treatment compared with mismatch control (p < 0.05, ANOVA). In addition, FACS analysis revealed a significant reduction in activated beta-catenin expression after imetelstat treatment of LSC engrafted mice compared with vehicle control (p < 0.01, ANOVA). Finally, RNA-seq analysis performed on human CD34+ cells from imetelstat treated LSC mouse models revealed a significant reduction in LSC harboring malignant ADAR1-mediated A-to-I editing at doses that spared normal hematopoietic stem cells.

CONCLUSIONS

Combined WGS and RNA-Seq analyses, lentiviral ADAR1 overexpression, stromal co-culture assays and humanized pre-LSC and LSC mouse model studies reveal that pre-LSC evolution into LSC coincides with both ADAR1 and hTERT activation, which can be prevented with imetelstat.

Disclosures: Rizo: Geron Corp: Current Employment, Current equity holder in publicly-traded company. Huang: Geron Corp: Current Employment, Current equity holder in publicly-traded company. Jamieson: Forty Seven Inc: Patents & Royalties; Bristol-Myers Squibb: Other.

*signifies non-member of ASH