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 #
5P50CA196530-05
Application #
9767072
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2019-08-01
Budget End
2020-07-31
Support Year
5
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Yale University
Department
Type
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06520
Altan, Mehmet; Kidwell, Kelley M; Pelekanou, Vasiliki et al. (2018) Association of B7-H4, PD-L1, and tumor infiltrating lymphocytes with outcomes in breast cancer. NPJ Breast Cancer 4:40
Kim, Tae Kon; Herbst, Roy S; Chen, Lieping (2018) Defining and Understanding Adaptive Resistance in Cancer Immunotherapy. Trends Immunol 39:624-631
Goldberg, Sarah B; Patel, Abhijit A (2018) Monitoring immunotherapy outcomes with circulating tumor DNA. Immunotherapy 10:1023-1025
Lopez Sambrooks, Cecilia; Baro, Marta; Quijano, Amanda et al. (2018) Oligosaccharyltransferase Inhibition Overcomes Therapeutic Resistance to EGFR Tyrosine Kinase Inhibitors. Cancer Res 78:5094-5106
Wilson, Frederick H; Politi, Katerina (2018) ERBB Signaling Interrupted: Targeting Ligand-Induced Pathway Activation. Cancer Discov 8:676-678
Wang, Guangchuan; Chow, Ryan D; Ye, Lupeng et al. (2018) Mapping a functional cancer genome atlas of tumor suppressors in mouse liver using AAV-CRISPR-mediated direct in vivo screening. Sci Adv 4:eaao5508
Villarroel-Espindola, Franz; Yu, Xiaoqing; Datar, Ila et al. (2018) Spatially Resolved and Quantitative Analysis of VISTA/PD-1H as a Novel Immunotherapy Target in Human Non-Small Cell Lung Cancer. Clin Cancer Res 24:1562-1573
Anastasiadou, Eleni; Jacob, Leni S; Slack, Frank J (2018) Non-coding RNA networks in cancer. Nat Rev Cancer 18:5-18
Bisserier, Malik; Wajapeyee, Narendra (2018) Mechanisms of resistance to EZH2 inhibitors in diffuse large B-cell lymphomas. Blood 131:2125-2137
Chow, Ryan D; Chen, Sidi (2018) Cancer CRISPR Screens In Vivo. Trends Cancer 4:349-358

Showing the most recent 10 out of 74 publications