The evasion of immune-mediated cytotoxicity is a hallmark feature of tumorigenesis. To this end, therapeutic inhibition of the immune checkpoint Programmed Cell Death Protein 1 (PD-1) has shown considerable promise in the management of several malignancies. In pancreatic cancer, however, despite a strong association between expression of the PD-1 ligand (PD-L1) and poor survival, checkpoint inhibitors have shown limited efficacy as a monotherapy. Recent efforts have therefore focused on identifying additional factors in the tumor microenvironment that modulate PD-L1/PD1 expression in hopes of improving drug response. Our group has previously identified stromal-derived Transforming Growth Factor Beta (TGF?) as a crucial repressor of anti- tumor immune function, particularly with respect to cytotoxic T lymphocytes. Despite these observations, pharmacologic inhibition of TGF? signaling has also been ineffective in clinical trials. When examining a small cohort of pancreatic cancer patients, we found an inverse association between PD-L1 expression and that of SMAD4, a downstream target of TGF? signals. In accordance with these observations, exogenous TGF?1 repressed the acquisition of PD-L1 in vitro. Similarly, neoplastic mouse models with genetic ablation of TGF? signaling developed increased PD-L1 expression in the pancreas, and failed to mount a full anti-tumor immune response. Using an established model of metastatic pancreatic cancer, we therefore administered pharmacologic inhibitors of PD-1 (Invivomab) and/or TGFBR1 (Galunisertib). While neither monotherapy had an effect, after two months, mice receiving both Invivomab and Galunisertib had no overt evidence of disease. Histologically, the pancreata of these mice had near complete regression of neoplasms, with abnormal tissues displaying increased lymphocyte infiltration, Granzyme deposition, and apoptosis. Combined, these data suggest that TGF? pathway blockade augments immune checkpoint inhibition, and may be a reasonable approach to overcome drug resistance in the clinic. Given the substantial preclinical efficacy of this approach, it is essential to further understand the means through which TGF? and PD-L1/PD-1 signals are regulated, as well as the merits to their inhibition in select patient populations. Through the experiments detailed in this proposal, we will gain valuable insight into these events in hopes of extending survival and reducing cancer- associated morbidity in PDAC patients.

Public Health Relevance

Pancreatic cancer has a dismal prognosis with a median survival time of 6-12 months, largely attributed to late diagnosis and multidrug resistance. It is therefore essential to understand the mechanisms by which these tumors acquire drug resistant phenotypes, particularly with respect to immune checkpoint inhibitors, which have shown remarkable efficacy in the management of other cancers. In this proposal, we outline a series of experiments dissecting the relationship between TGF? signaling and the immune checkpoint molecule PD-L1, as well as those evaluating the potential therapeutic efficacy of concomitant TGF?/PD-L1 pathway inhibition in hopes of enhancing anti-tumor cytotoxicity and improving patient survival.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Individual Predoctoral NRSA for M.D./Ph.D. Fellowships (ADAMHA) (F30)
Project #
5F30CA236031-03
Application #
10067527
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Bian, Yansong
Project Start
2019-01-02
Project End
2024-01-01
Budget Start
2021-01-02
Budget End
2022-01-01
Support Year
3
Fiscal Year
2021
Total Cost
Indirect Cost
Name
University of Illinois at Chicago
Department
Surgery
Type
Schools of Medicine
DUNS #
098987217
City
Chicago
State
IL
Country
United States
Zip Code
60612