-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.

4269 In Vivo Response to Cytarabine Chemotherapy Uncovers the Role of the Oxidative and Energetic Metabolism in the Chemoresistance of Human Primary AML Stem Cells

Malignant Stem and Progenitor Cells
Program: Oral and Poster Abstracts
Session: 661. Malignant Stem and Progenitor Cells: Poster III
Monday, December 7, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Nesrine Aroua, PhD1*, Jean-Emmanuel Sarry, PhD2*, Fabienne De Toni, PhD3*, Robin Carson, MD, BA4, Francois Vergez, VD PhD5*, Estelle Saland2*, Mayumi Sugita, MD6*, Martin Carroll, MD7, Christian Recher, MD-Ph.D.8 and Gwenn Danet, PhD9*

1INSERM, CRCT-U1037, Toulouse, France
2INSERM, CRCT, Toulouse, France
3CRCT, INSERM, Toulouse, France
4Janssen Research & Development, Spring House, PA
5Institut Universitaire du Cancer, Toulouse, France
6Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA
7Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
8Institut Universitaire du Cancer de Toulouse, Toulouse, France
9Univ. of Chicago Medical Center, Chicago, IL

Resistant cells to conventional chemotherapy - which targets DNA synthesis - are thought to be rare and enriched in quiescent leukemic stem cells (LSCs) in acute myeloid leukemia (AML). In order to develop an improved understanding of chemotherapy resistance in AML, we analyzed the response to cytarabine (Ara-C) in our xenograft model with 20 primary AML patient specimens from two clinical sites. After demonstrating AML engraftment, mice are treated with Ara-C given IP daily for 5 days as a single agent at 60 mg/kg daily, which correlates with human dosing. In all mice treated with this regimen, there was a 4- to 46-fold cytoreductive effect at 1 week post-treatment. While in vivo Ara-C treatment induces changes in CD34posCD38pos/neg phenotypes of 50% of AML-engrafted mice, this treatment does not induce any significant changes in cell cycle distribution and does not enrich in G0 quiescent cells in AML-engrafted mice. Surprisingly we observed that Ara-C induces a decrease or no change in the frequency of SL-ICs (control, 1:2,500-21,900 versus Ara-C, 1:4,500-34,200), suggesting that Ara-C would not spare LSCs in vivo and equally kills both cycling and quiescent cells. Furthermore, transcriptomic analysis of Ara-C residual leukemic cells (RLCs) from three different patient-derived xenografts confirmed that RLCs are not enriched in stem cell marker genes at 1 week post-treatment. However, our gene signature of the in vivo AML resistance is enriched in genes involved in inflammatory, immune and stress response. When tested in three independent AML cohorts (Verhaak et al. 2009; TGCA, 2011; Metzeler et al. 2011), this signature is associated with an unfavorable prognosis in patients treated with intensive chemotherapy. Finally we have shown that RLCs are PIMOpos cells with both high ROS content and active mitochondrial mass and membrane potential in AraC-treated xenografted mice compared to control mice. Accordingly, AML cells exhibiting high OXPHOS energetic phenotype are resistant to Ara-C chemotherapy in vivo while low OXPHOS AML cells are sensitive to AraC in NSG mice. Altogether these results suggest a novel model of cytarabine chemotherapy resistance in AML based on the oxidative and mitochondrial energy metabolism in vivo.

Disclosures: No relevant conflicts of interest to declare.

*signifies non-member of ASH