-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.

3659 Immunomodulatory Effect of MUC1-C in Acute Myeloid Leukemia

Oncogenes and Tumor Suppressors
Program: Oral and Poster Abstracts
Session: 603. Oncogenes and Tumor Suppressors: Poster III
Monday, December 7, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Dina Stroopinsky, PhD1*, Hasan Rajabi, PhD2*, Maxwell Douglas Coll1*, Athalia Rachel Pyzer, MD1*, Jacalyn Rosenblatt, MD1, Salvia Jain, MD3, Katarina Luptakova, MD1, Malgorzata McMasters, MD1*, James D. Levine, MD1, Robin Joyce, MD4, Jon E. Arnason, MD1, Rebecca Karp Leaf, MD1, Myrna Rita Nahas, MD1*, Kristen Anna Palmer1*, Michal Bar-Natan, MD1*, Ashujit Tagde, PhD, BPharm2*, Abigail Washington1*, Poorvi Somaiya1*, Donald Kufe, MD2* and David E. Avigan, MD1

1Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
2Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
3Malignant Hematology and Bone Marrow Transplantation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
4Division of Hematology and Oncology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA

Acute myeloid leukemia (AML) is characterized by an immunosuppressive milieu in the bone marrow microenvironment that fosters tolerance, immune escape, and disease growth. Upregulation of PD-L1 expression by leukemia cells induces an exhausted phenotype in tumor reactive lymphocytes that blocks the activation and expansion of leukemia specific lymphocytes.  Identifying pathways that regulate PD-L1 expression is vital for the development of novel therapeutic targets for immune based therapies.  MUC1 is an oncoprotein that is aberrantly expressed on a majority of primary AML. MUC1 contains a transmembrane subunit (MUC1-C) that forms dimers and translocates to the nucleus where it mediates downstream signaling. MUC1-C expression on malignant cells promotes cell proliferation and resistance to apoptosis.

We have demonstrated that MUC1-C is a critical regulator of PD-L1 expression on AML cells.  Silencing of MUC1 expression was documented following lentiviral transduction with MUC1-C specific shRNA using western blotting.  MUC1 silencing results in the near abrogation of MUC1-C expression by the human AML cell lines, MOLM-14 and THP1. Silencing of MUC1-C on MOLM-14 and THP1 AML cells resulted in a two-fold increase in susceptibility to T cell mediated lysis as determined by a flourochrome based CTL assay.

To assess the effect of MUC1-C silencing on PD-L1 expression in-vivo, C57BL/6J mice were challenged with 100,000 GFP transfected TIB-49 murine AML cells in which MUC1-C was silenced using a lentiviral shRNA hairpin against MUC1-C.   Following leukemia establishment, TIB-49 GFP+ cells were isolated from the bone marrows and spleens and PD-L1 expression on leukemic cells was measured. TIB-49 GFP+ cells of mice engrafted with MUC1 silenced AML cells demonstrated significantly lower PD-L1 expression compared to mice inoculated with TIB-49 cells transduced with a control vector (18% versus 3%; n=4).  T cells isolated from bone marrow of mice inoculated with AML with silenced MUC1-C demonstrated a three-fold increase in INF-γ production when stimulated ex-vivo with autologous tumor lysate as compared to T cells from mice inoculated with control AML cells (n=4).  Our group has developed a peptide inhibitor drug (G0-203), a cell-penetrating peptide that binds to the MUC1-C CQC motif, disrupting MUC1-C interaction with downstream effectors. As was observed following MUC1 silencing, bone marrow derived CD4+ and CD8+ T cells isolated from mice treated with G0-203 were shown to have two-fold increased intracellular IFN-γ production compared to control mice following ex-vivo exposure to AML lysate (n=4).

To investigate the mechanism by which MUC1-C regulates PD-L1 expression we examined the role of noncoding RNAs that have been shown to be effectors of oncogenic modulation. MicroRNAs (miRNAs) are a conserved class of small RNAs that post-transcriptionally regulate gene expression by interacting with the 3′ untranslated region (3′ UTR) of target mRNAs. The 3’UTR of the PD-L1 gene contains putative binding sites for miR-200 family of micro-RNAs, supporting the hypothesis that miR-200 plays a role in regulating the expression of PD-L1.

In the present study, we evaluated the effect of targeting MUC1-C mediated signaling, on miR200c levels and PD-L1 expression. MUC1-C silenced MOLM-14 cells demonstrated a two-fold increase in miR-200c expression, as demonstrated by qPCR and was associated with a reduction of PD-L1 expression from 77% to 13%. These data were confirmed in THP1 cells with PDL-1 expression decreasing from 95% to 40% following MUC1-C silencing. In support of the observation that miR200c is involved in the regulation of PD-L1 expression, lentiviral overexpression of miR200c in MOLM-14 cells led to a decrease in PD-L1 expression from 90% to 2% as demonstrated by flow cytometric analysis.

In conclusion, these results demonstrate that MUC1-C mediated signaling regulates the expression of PD-L1 on AML cells.  Silencing MUC1-C results in reduced expression of PD-L1, interfering with immune escape and enhancing the expansion of leukemia reactive T cells. We define a novel mechanism by which MUC1-C governs PD-L1 expression through its modulation of miR200c.  As such, MUC1-C functions as a critical regulator of immune escape and represents a novel target for immunotherapy.

Disclosures: Kufe: Genus Oncology: Consultancy , Equity Ownership . Avigan: Regimmune: Research Funding .

*signifies non-member of ASH