Recent evidence suggests that tumors may exploit immune inhibitory mechanisms to create a barrier against antitumor immune responses ? including endogenous inflammatory immune responses and those induced by immunotherapies. One key mechanism appears to be mediated by the programmed death-1 (PD-1)/B7 homolog 1 (B7-H1) pathway, which restrains antitumor T cell function in the tumor microenvironment (TME). PD-1 is induced on the surface of activated T cells and, after engaging its ligand, down-modulates T cell functions. Our early studies include cloning and identification of B7-H1 (PD-L1, CD274) as an inhibitory ligand for T cell response and characterization of B7-H1 as a major ligand for PD-1 in suppressing tumor immunity in TME. We demonstrated that B7-H1 was upregulated on the cell surface by inflammation (mainly via IFN-gamma), and that high levels of B7-H1 expression were found in many human cancers including non-small cell lung cancer (NSCLC). Blocking the interaction of B7-H1 on tumor cells with PD-1 on tumor-specific T cells can eliminate this barrier in the TME and protect ongoing antitumor immunity, leading to tumor regression in mouse tumor models. Recent clinical trials have demonstrated that anti-PD-1 or anti-B7-H1 monoclonal antibodies (mAb) induced objective clinical responses in a significant fraction of patients with advanced chemo-refractory metastatic NSCLC. Responses were highly durable with manageable toxicity. While these clinical findings are promising, it is now critical to better understand the effects of PD-1/B7-H1 blockade on antitumor immunity and to develop new strategies to overcome resistance. In this project, we will test the hypothesis that NSCLC is immunologically heterogeneous and only a subgroup of NSCLC with membranous B7-H1 (PD-L1) expression on cancer cells and the presence of functional tumor-infiltrating lymphocytes (TILs) will respond to anti-PD- 1/B7-H1 therapy, whereas resistance to anti-PD-1/B7-H1 therapy is largely mediated via PD-1/B7-H1- independent suppressive pathways. We will identify subgroups of NSCLC patients who respond to and resist anti-PD-1/B7-H1 therapy by analysis of B7-H1 expression and immune responses in the TME. In addition, we will delineate cellular and molecular mechanisms underlying resistance to anti-PD-1/B7-H1 therapy in each NSCLC subgroup. Finally, we will maximize the therapeutic effect of PD-1/B7-H1 blockade by additionally targeting resistance mechanisms using mouse models with induced lung cancer. The current proposal integrates basic and clinical sciences, and will use animal models and human specimens in the context of ongoing clinical trials of anti-PD-1 and B7-H1 antibodies to achieve the goals. Taken together, results from our studies will enhance future anti-PD-1/B7-H1 therapy and potentially lead to novel immune-based therapies for lung cancer.

Public Health Relevance

Despite advances in therapeutics for NSCLC, the need for more effective, better tolerated therapies is clear. Recent clinical trials evaluating anti-PD-1/B7-H1 therapy have shown promising clinical responses and survival benefit in some patients, with less overall toxicity compared to standard anti-tumor therapies. This proposal aims to better understand both sensitivity and resistance to such therapy, to enable selection of anti-PD-1/B7- H1 therapy for patients most likely to respond, and to develop therapeutics to overcome resistance to PD-1/ B7-H1 blockade.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
5P50CA196530-03
Application #
9325321
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2017-08-01
Budget End
2018-07-31
Support Year
3
Fiscal Year
2017
Total Cost
Indirect Cost
Name
Yale University
Department
Type
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06520
Gilles, Maud-Emmanuelle; Slack, Frank J (2018) Let-7 microRNA as a potential therapeutic target with implications for immunotherapy. Expert Opin Ther Targets 22:929-939
Nagarajan, Maxwell B; Tentori, Augusto M; Zhang, Wen Cai et al. (2018) Nonfouling, Encoded Hydrogel Microparticles for Multiplex MicroRNA Profiling Directly from Formalin-Fixed, Paraffin-Embedded Tissue. Anal Chem 90:10279-10285
Hartman, Douglas J; Ahmad, Fahad; Ferris, Robert L et al. (2018) Utility of CD8 score by automated quantitative image analysis in head and neck squamous cell carcinoma. Oral Oncol 86:278-287
Nagarajan, Arvindhan; Malvi, Parmanand; Wajapeyee, Narendra (2018) Heparan Sulfate and Heparan Sulfate Proteoglycans in Cancer Initiation and Progression. Front Endocrinol (Lausanne) 9:483
Rojewski, Alana M; Tanner, Nichole T; Dai, Lin et al. (2018) Tobacco Dependence Predicts Higher Lung Cancer and Mortality Rates and Lower Rates of Smoking Cessation in the National Lung Screening Trial. Chest 154:110-118
Chen, Ling; Azuma, Takeshi; Yu, Weiwei et al. (2018) B7-H1 maintains the polyclonal T cell response by protecting dendritic cells from cytotoxic T lymphocyte destruction. Proc Natl Acad Sci U S A 115:3126-3131
Zhang, Jinhua; Song, Kun; Wang, Jun et al. (2018) S100A4 blockage alleviates agonistic anti-CD137 antibody-induced liver pathology without disruption of antitumor immunity. Oncoimmunology 7:e1296996
Anastasiadou, Eleni; Faggioni, Alberto; Trivedi, Pankaj et al. (2018) The Nefarious Nexus of Noncoding RNAs in Cancer. Int J Mol Sci 19:
Toki, Maria I; Mani, Nikita; Smithy, James W et al. (2018) Immune Marker Profiling and Programmed Death Ligand 1 Expression Across NSCLC Mutations. J Thorac Oncol 13:1884-1896
Park, Seyoung; Zhao, Hongyu (2018) Spectral clustering based on learning similarity matrix. Bioinformatics 34:2069-2076

Showing the most recent 10 out of 74 publications