Immunotherapy with PD-1/PD-L1 checkpoint blockade (PCB) given alone or with chemotherapy now represents the standard first-line treatment for NSCLC patients with wild-type (wt) EGFR and ALK. This is a major advance, but the majority of NSCLC patients do not have an objective response to PCB. The molecular determinants of PCB resistance are incompletely understood, although low tumor mutation burden and PD-L1 levels predict some cases. Recently, we reported that LKB1 deficient (LD) tumors resulting from mutations or deletions in the STK11/LKB1 gene are associated with an inert or ?cold? immune phenotype and represent the largest genomically-define subgroup with primary resistance to PCB, accounting for more than 30% of PCB resistance in lung adenocarcinoma. The LKB1 protein is a master regulator of metabolism, energetic balance, and nucleotide stores. Recent publications from our group and others indicate that LD tumors have a distinct metabolic phenotype that includes enhanced lactate production, vulnerability to targeting intracellular nucleotide pathways, and increased replicative stress (RS). These features may contribute to the ?cold? immune phenotype. The primary goals of Project 2 (P2) are to investigate new therapeutic approaches for targeting LD NSCLC and enhancing antitumor immunity, with a focus on targeting the lactate pathway and RS. This focus integrates the immunotherapy focus of Project 2 with studies in the other SPORE Projects and provides multiple Human Endpoints for the SPORE Projects. We hypothesize that: 1) Enhanced lactate production or secretion contributes to the ?cold? immune phenotype in LD NSCLC; 2) LD NSCLC will be preferentially vulnerable to targeting RS; and 3) targeting RS and/or lactate pathways may enhance antitumor immunity and response to PCB. To test these hypotheses, in SA1 we will comprehensively characterize the immune phenotypes of LD NSCLC using two sets of resected tumors: the MD Anderson ICON cohort and a validation cohort from UTSW which have undergone metabolic labeling in P1. We will also examine immune cells in greater detail using single cell RNA sequencing. In SA2 we will test whether targeting the lactate pathway using inhibitors of MCT4 and LDHA can reverse LD-associated immunosuppression and enhance PCB efficacy. In SA3, we will target RS using inhibitors of ATM, ATR, and the nucleoside analog 6-thio-dG in collaboration with P4, alone or in combination with PCB. Significance: LD NSCLC tumors have a ?cold? immune phenotype and frequent primary resistance to PCB or PCB/chemotherapy. This patient population is larger than EGFR-mutant NSCLC and comparably sized to metastatic pancreatic cancer.
P2 aims to leverage our unique set of clinical and preclinical resources to develop more effective therapeutic approaches for LD NSCLC patients, which can then translated by our group and others into the clinic. P2 also provides the opportunity to spearhead a new paradigm of genomically-guided, tailored immunotherapy for PCB-resistant tumors.

Public Health Relevance

Although PD-1/PD-L1 immune checkpoint blockade (PCB) therapy has transformed the treatment of non-small cell lung cancer (NSCLC) patients, the majority still do not have an objective response to PCB and the molecular determinants of therapeutic resistance are not well understood. We have found that STK11/LKB1 mutant NSCLC is one the largest genomic subgroups associated with a ?cold? immune phenotype, accounting for ~40% of the primary resistance to checkpoint blockade therapy, and we hypothesize that there are distinct biological features of these tumors that may contribute their immunologically inert phenotype and that represent therapeutic vulnerabilities. In this proposal will leverage unique preclinical and clinical resources to comprehensively investigate the immune landscape of STK11/LKB1 mutant lung adenocarcinoma and test whether targeting two potential therapeutic vulnerabilities- lactate metabolism and replication stress- can enhance antitumor immunity in these tumors.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
2P50CA070907-21A1
Application #
10023866
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2020-09-01
Budget End
2021-08-31
Support Year
21
Fiscal Year
2020
Total Cost
Indirect Cost
Name
University of Texas Sw Medical Center Dallas
Department
Type
DUNS #
800771545
City
Dallas
State
TX
Country
United States
Zip Code
75390
He, Min; Liu, Shanshan; Gallolu Kankanamalage, Sachith et al. (2018) The Epithelial Sodium Channel (?ENaC) Is a Downstream Therapeutic Target of ASCL1 in Pulmonary Neuroendocrine Tumors. Transl Oncol 11:292-299
Parra, Edwin R; Villalobos, Pamela; Behrens, Carmen et al. (2018) Effect of neoadjuvant chemotherapy on the immune microenvironment in non-small cell lung carcinomas as determined by multiplex immunofluorescence and image analysis approaches. J Immunother Cancer 6:48
Guo, Hou-Fu; Tsai, Chi-Lin; Terajima, Masahiko et al. (2018) Pro-metastatic collagen lysyl hydroxylase dimer assemblies stabilized by Fe2+-binding. Nat Commun 9:512
Meraz, Ismail M; Majidi, Mourad; Cao, Xiaobo et al. (2018) TUSC2 Immunogene Therapy Synergizes with Anti-PD-1 through Enhanced Proliferation and Infiltration of Natural Killer Cells in Syngeneic Kras-Mutant Mouse Lung Cancer Models. Cancer Immunol Res 6:163-177
Zhang, Liren; Lin, Jing; Ye, Yuanqing et al. (2018) Serum MicroRNA-150 Predicts Prognosis for Early-Stage Non-Small Cell Lung Cancer and Promotes Tumor Cell Proliferation by Targeting Tumor Suppressor Gene SRCIN1. Clin Pharmacol Ther 103:1061-1073
Bayo, Juan; Tran, Tram Anh; Wang, Lei et al. (2018) Jumonji Inhibitors Overcome Radioresistance in Cancer through Changes in H3K4 Methylation at Double-Strand Breaks. Cell Rep 25:1040-1050.e5
Ludlow, Andrew T; Wong, Mandy Sze; Robin, Jerome D et al. (2018) NOVA1 regulates hTERT splicing and cell growth in non-small cell lung cancer. Nat Commun 9:3112
Chen, Limo; Diao, Lixia; Yang, Yongbin et al. (2018) CD38-Mediated Immunosuppression as a Mechanism of Tumor Cell Escape from PD-1/PD-L1 Blockade. Cancer Discov 8:1156-1175
Mender, Ilgen; LaRanger, Ryan; Luitel, Krishna et al. (2018) Telomerase-Mediated Strategy for Overcoming Non-Small Cell Lung Cancer Targeted Therapy and Chemotherapy Resistance. Neoplasia 20:826-837
Gong, Ke; Guo, Gao; Gerber, David E et al. (2018) TNF-driven adaptive response mediates resistance to EGFR inhibition in lung cancer. J Clin Invest 128:2500-2518

Showing the most recent 10 out of 1059 publications