-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.

443 Targeting MTHFD2 in Acute Myeloid Leukemia

Oncogenes and Tumor Suppressors
Program: Oral and Poster Abstracts
Type: Oral
Session: 603. Oncogenes and Tumor Suppressors: Post-Transcriptional and Metabolic Mechanisms of Leukemogenesis
Monday, December 7, 2015: 8:00 AM
W308, Level 3 (Orange County Convention Center)

Yana Pikman, MD1, Alexandre Puissant, PhD2*, Gabriela Alexe, PhD3*, Andrew Furman, BA3*, Stacey Frumm, BA4, Linda Ross, PhD3*, Liying Chen, PhD3*, Nina Fenouille, PhD5*, Christopher F. Bassil, BA3*, Caroline A Lewis, PhD5*, Richard M. Stone, MD6, Daniel J. DeAngelo, MD, PhD7, Ilene Galinsky, ANP7, Andrew L Kung, MD, PhD8, Matthew Vader Heiden, MD, PhD5*, Michael T. Hemann, PhD5*, Versha Banerji, MD9 and Kimberly Stegmaier, MD1

1Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA
2Department of Pediatric Oncology, Dana-Farber Cancer Instituté, Boston, MA
3Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
4UCSF, San Francisco, CA
5Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA
6Hematologic Oncology, Dana Farber Cancer Institute, Boston, MA
7Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
8Division of Pediatric Hematology/Oncology/Stem Cell Transplantation, Columbia University Medical Center, New York, NY
9Research Institute of Oncology and Hematology, Cancer Care Manitoba, Winnipeg, MB, Canada

Alterations in differentiation pathways contribute to the development of acute myeloid leukemia (AML). Differentiation therapy with all-trans retinoic acid (ATRA) has dramatically altered the treatment of acute promyelocytic leukemia, transforming it from a nearly fatal disease to a curable one. We set out to identify cellular pathways that contribute to AML differentiation, with the goal of identifying new therapeutic targets. We analyzed gene expression data from AML cell lines treated with phorbol 12-myristate 13-acetate (PMA), ATRA, Vitamin D, the BET inhibitor JQ1 and the DOT1L inhibitor EPZ00477­, treatments known to induce AML differentiation and impair growth. Folate-mediated one-carbon metabolism was one of only three metabolic pathways altered by these compounds, with expression of MTHFD2 consistently downregulated with each compound.

MTHFD2 is an NAD-dependent, bi-functional mitochondrial methylenetetrahydrofolate dehydrogenase and cyclohydrolase. It is differentially expressed in embryonic and transformed tissues and is upregulated in myeloid progenitors. MTHFD2 is the most differentially expressed metabolic enzyme in cancer cells versus normal cells, including normal proliferating cells. We thus investigated the role of MTHFD2 in myeloid malignancy. First, we demonstrated using ChIP-qPCR, MYC knockdown and MYC inhibition with a BET inhibitor, that MYC directly regulates MTHFD2 expression in AML. Knockdown of MTHFD2 with two shRNAs confirmed to have on-target activity, induced myeloid differentiation in AML cell lines, as measured by Cd11b expression, morphologic changes and induction of a previously validated AML differentiation gene expression signature. MTHFD2 knockdown decreased cell growth in AML cell lines, as well as decreased colony formation in methylcellulose in both AML cell lines and primary patient blasts. AML cells transduced with these two MTHFD2-directed shRNAs demonstrated attenuated growth in an orthotopic mouse model of AML. Furthermore, three MTHFD2-directed shRNAs prolonged survival in an MLL-AF9 mouse leukemia model. Additionally, using a doxycycline inducible shRNA system, we demonstrated that two miR30-shRNAs directed against MTHFD2 decreased AML burden in mice with established disease and prolonged survival.

To identify biomarkers of response to MTHFD2 suppression, we used single sample Gene Set Enrichment Analysis (ssGSEA) to identify primary patient AML samples enriched for gene expression signatures of folate-mediated one-carbon metabolism and MTHFD2. We found in both independent data sets that the cluster of patients enriched for expression of the one-carbon folate pathway gene signatures was also enriched for patients with FLT3-ITD mutations, a subset of AML with a particularly poor prognosis. In addition, in an shRNA screen targeting 11,194 genes and performed in 216 cancer cell lines, including 17 AML lines, FLT3-ITD was a biomarker of response to MTHFD2 knockdown. We next validated that while MTHFD2 suppression induced measureable differentiation in all six AML cell lines examined, it induced the most robust induction of apoptosis in FLT3-ITD mutant AML. The mitochondrial one-carbon folate pathway is thought to contribute to cellular oxidative balance by providing reducing power in the form of NAD(P)H, and suppression of MTHFD2 was previously shown to increase ROS levels. Indeed, suppression of MTHFD2 led to a marked increase in ROS in the FLT3-ITD positive AML cell lines in which apoptosis was induced.

In summary, a decrement in MTHFD2 expression was found at the center of multiple AML perturbations that impair AML growth and induce differentiation. Our data support MTHFD2 as an AML dependency and FLT3-ITD as a potential biomarker of response. We thus provide critical preclinical evidence for targeting of MTHFD2 as a therapeutic strategy in AML.

Disclosures: Stone: Celgene: Consultancy ; Merck: Consultancy . DeAngelo: Celgene: Consultancy ; Pfizer: Consultancy ; Incyte: Consultancy ; Agios: Consultancy ; Novartis: Consultancy ; Ariad: Consultancy ; Bristol Myers Squibb: Consultancy ; Amgen: Consultancy . Stegmaier: Novartis Pharmaceuticals: Consultancy .

*signifies non-member of ASH