Mucin 1 (MUC1) is aberrantly overexpressed in ~90% of human breast cancers, including the triple-negative (TNBC) subtype, and is associated with poor progression-free and overall survival. MUC1 has thus emerged as a highly attractive target for the treatment of TNBC; however, to date there are no approved agents against this heterodimeric transmembrane protein. What is needed now, at least in part, is therefore the development of novel agents that target MUC1-C for the treatment of patients with refractory TNBC. Importantly, MUC1 consists of two subunits: an extracellular N-terminal mucin subunit (MUC1-N) that is shed from the cell surface, and a transmembrane C-terminal subunit (MUC1-C) that is oncogenic. MUC1-C functions as an oncoprotein by acting as a node for integrating signaling pathways linked to transformation. In this way, MUC1-C drives (i) the epithelial-mesenchymal transition (EMT), (ii) self-renewal capacity, and (iii) tumorigenicity of TNBC cells. Early attempts at targeting MUC1-N with monoclonal antibodies (MAbs) were unsuccessful because this shed subunit circulates at high levels in the blood of women with breast cancer. As an alternative approach, we have generated novel MAbs against the non-shed MUC1-C extracellular domain. These MAbs have provided a unique opportunity to develop antibody-drug conjugates (ADCs) that specifically target MUC1-C on the surface of breast cancer cells. Our MAbs are also being advanced for the development of antibody-dependent cell-mediated cytotoxicity (ADCC) and for combinations with other immunotherapies. Recent Phase I trials using single-agent anti-PD-1 and anti-PD-L1 blockade have demonstrated objective and durable responses in heavily pretreated, metastatic TNBC, supporting the premise that evasion of immune destruction contributes to TNBC pathogenesis. Our hypothesis is that our ADCs will be effective against MUC1-expressing breast tumors in our immunocompetent mouse models and that killing these tumor cells could reverse, at least in part, the immune suppressive effects of the tumor microenvironment. In this R21 application, our immunotherapeutic approaches targeting MUC1-C will be developed for clinical evaluation based on their effectiveness against TNBC cells growing in vitro and in animal models. The overall objective of the proposed work is thus to improve the immunotherapy of breast cancer. Our hypothesis is that MUC1-C is an attractive target and that our immunotherapeutic approaches can be developed for patients with TNBC.

Public Health Relevance

Breast cancer is one of the leading causes of death for women. The MUC1 oncogenic protein is aberrantly overexpressed in about 90% of human breast tumors and confers a poor survival outcome for patients. Our proposed research focuses on how to effectively target MUC1 function in breast cancer with the novel immunotherapeutic approaches we have developed for the treatment of this disease.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Exploratory/Developmental Grants (R21)
Project #
5R21CA216553-02
Application #
9455644
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Welch, Anthony R
Project Start
2017-03-16
Project End
2019-02-28
Budget Start
2018-03-01
Budget End
2019-02-28
Support Year
2
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Dana-Farber Cancer Institute
Department
Type
DUNS #
076580745
City
Boston
State
MA
Country
United States
Zip Code
Rajabi, Hasan; Hiraki, Masayuki; Kufe, Donald (2018) MUC1-C activates polycomb repressive complexes and downregulates tumor suppressor genes in human cancer cells. Oncogene 37:2079-2088
Panchamoorthy, Govind; Jin, Caining; Raina, Deepak et al. (2018) Targeting the human MUC1-C oncoprotein with an antibody-drug conjugate. JCI Insight 3:
Hiraki, Masayuki; Maeda, Takahiro; Mehrotra, Neha et al. (2018) Targeting MUC1-C suppresses BCL2A1 in triple-negative breast cancer. Signal Transduct Target Ther 3:13
Maeda, Takahiro; Hiraki, Masayuki; Jin, Caining et al. (2018) MUC1-C Induces PD-L1 and Immune Evasion in Triple-Negative Breast Cancer. Cancer Res 78:205-215
Bouillez, Audrey; Adeegbe, Dennis; Jin, Caining et al. (2017) MUC1-C promotes the suppressive immune microenvironment in non-small cell lung cancer. Oncoimmunology 6:e1338998
Bouillez, A; Rajabi, H; Jin, C et al. (2017) MUC1-C integrates PD-L1 induction with repression of immune effectors in non-small-cell lung cancer. Oncogene 36:4037-4046
Jain, Salvia; Washington, Abigail; Leaf, Rebecca Karp et al. (2017) Decitabine Priming Enhances Mucin 1 Inhibition Mediated Disruption of Redox Homeostasis in Cutaneous T-Cell Lymphoma. Mol Cancer Ther 16:2304-2314
Rajabi, Hasan; Hiraki, Masayuki; Tagde, Ashujit et al. (2017) MUC1-C activates EZH2 expression and function in human cancer cells. Sci Rep 7:7481