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 #
1P50CA196530-01
Application #
8931833
Study Section
Special Emphasis Panel (ZCA1-RPRB-7 (M1))
Project Start
2015-08-26
Project End
2020-07-31
Budget Start
2015-08-26
Budget End
2016-07-31
Support Year
1
Fiscal Year
2015
Total Cost
$197,114
Indirect Cost
Name
Yale University
Department
Microbiology/Immun/Virology
Type
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06510
Burslem, George M; Smith, Blake E; Lai, Ashton C et al. (2018) The Advantages of Targeted Protein Degradation Over Inhibition: An RTK Case Study. Cell Chem Biol 25:67-77.e3
Liu, Huafeng; Li, Xin; Hu, Li et al. (2018) A crucial role of the PD-1H coinhibitory receptor in suppressing experimental asthma. Cell Mol Immunol 15:838-845
Choe, Junho; Lin, Shuibin; Zhang, Wencai et al. (2018) mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis. Nature 561:556-560
Bade, Brett C; Hyer, J Madison; Bevill, Benjamin T et al. (2018) A Patient-Centered Activity Regimen Improves Participation in Physical Activity Interventions in Advanced-Stage Lung Cancer. Integr Cancer Ther 17:921-927
Tentori, Augusto M; Nagarajan, Maxwell B; Kim, Jae Jung et al. (2018) Quantitative and multiplex microRNA assays from unprocessed cells in isolated nanoliter well arrays. Lab Chip 18:2410-2424
Fan, Pang-Dian; Narzisi, Giuseppe; Jayaprakash, Anitha D et al. (2018) YES1 amplification is a mechanism of acquired resistance to EGFR inhibitors identified by transposon mutagenesis and clinical genomics. Proc Natl Acad Sci U S A 115:E6030-E6038
Gupta, Swati; Mani, Navin R; Carvajal-Hausdorf, Daniel E et al. (2018) Macrodissection prior to closed system RT-qPCR is not necessary for estrogen receptor and HER2 concordance with IHC/FISH in breast cancer. Lab Invest 98:1076-1083
Robichaux, Jacqulyne P; Elamin, Yasir Y; Tan, Zhi et al. (2018) Mechanisms and clinical activity of an EGFR and HER2 exon 20-selective kinase inhibitor in non-small cell lung cancer. Nat Med 24:638-646
Bondeson, Daniel P; Smith, Blake E; Burslem, George M et al. (2018) Lessons in PROTAC Design from Selective Degradation with a Promiscuous Warhead. Cell Chem Biol 25:78-87.e5
O'Malley, Stephanie S; Zweben, Allen; Fucito, Lisa M et al. (2018) Effect of Varenicline Combined With Medical Management on Alcohol Use Disorder With Comorbid Cigarette Smoking: A Randomized Clinical Trial. JAMA Psychiatry 75:129-138

Showing the most recent 10 out of 74 publications