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. Therefore, what is needed now is the development of novel agents that target MUC1 and specifically the MUC1-C subunit for the treatment of patients with refractory TNBC. 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) the cancer stem cell (CSC) state, (iii) tumorigenicity, (iv) metabolic alterations, (v) epigenetic programming, and (vi) immune evasion of TNBC cells. Our work has also demonstrated that MUC1-C is a druggable target. As one approach, we have generated first-in-class monoclonal antibodies (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 TNBC cells. Our MAbs are also being advanced for the development of antibody- dependent cell-mediated cytotoxicity (ADCC) and bispecific immunotherapeutics. In addition, we have developed agents that target the MUC1-C cytoplasmic domain and inhibit its oncogenic function. Targeting MUC1-C with the GO-203 inhibitor reverses the EMT, CSC and tumorigenic TNBC phenotype. Targeting MUC1-C with GO-203 also inhibits immune evasion of TNBC cells, indicating that GO- 203 could be used in combination with other immunotherapies. These findings and the development of highly novel antibodies have collectively supported the targeting of MUC1-C as potential immunotherapeutic approaches for patients with TNBC.
The Specific Aims are: (1) To investigate the effects of ADCs targeting the MUC1-C extracellular domain on TNBC cells; (2) To develop an ADCC and a bispecific antibody for targeting MUC1-C on TNBCs; (3) To target the MUC1-C cytoplasmic domain with GO-203/NPs in combination with other immunotherapeutics to circumvent TNBC immune evasion; and (4) To analyze TNBC specimens for MUC1-C expression and the suppressive immune microenvironment as metrics for response to MUC1-C-targeted agents.

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 the aggressive triple-negative subtype of breast cancer with the novel agents we have developed for the treatment of this disease.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA097098-17
Application #
9933812
Study Section
Developmental Therapeutics Study Section (DT)
Program Officer
Chen, Weiwei
Project Start
2002-07-01
Project End
2023-05-31
Budget Start
2020-06-01
Budget End
2021-05-31
Support Year
17
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Dana-Farber Cancer Institute
Department
Type
DUNS #
076580745
City
Boston
State
MA
Country
United States
Zip Code
02215
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:
Stroopinsky, Dina; Rajabi, Hasan; Nahas, Myrna et al. (2018) MUC1-C drives myeloid leukaemogenesis and resistance to treatment by a survivin-mediated mechanism. J Cell Mol Med :
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
Gupta, Dikshi; Kumar, Manoj; Tyagi, Priyanka et al. (2018) Concomitant Delivery of Paclitaxel and NuBCP-9 peptide for synergistic enhancement of cancer therapy. Nanomedicine 14:1301-1313
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
Hiraki, M; Maeda, T; Bouillez, A et al. (2017) MUC1-C activates BMI1 in human cancer cells. Oncogene 36:2791-2801
Yin, Li; Tagde, Ashujit; Gali, Reddy et al. (2017) MUC1-C is a target in lenalidomide resistant multiple myeloma. Br J Haematol 178:914-926
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
Shukla, Vasundhara; Dalela, Manu; Vij, Manika et al. (2017) Systemic delivery of the tumor necrosis factor gene to tumors by a novel dual DNA-nanocomplex in a nanoparticle system. Nanomedicine 13:1833-1839

Showing the most recent 10 out of 81 publications