The goal of this research project proposal is to co-opt dysfunctional innate immune signaling in KRAS- driven lung cancers as a therapeutic vulnerability. Specifically, my laboratory has focused for many years on targeting TBK1 to inhibit the production of cytokines and chemokines such as IL-6 and CCL5 that are pro- tumorigenic and immune suppressive. For example, TBK1 inhibition sensitizes tumors to MEK inhibition, as well as PD-1 blockade. Over the past several years it has also become increasingly apparent that viral sensing pathways, such as RIG-I/MAVS or cGAS/STING play a key role in re-directing TBK1 to activate IRF3/STAT1 and initiate a cytotoxic anti-viral response. How KRAS mutant lung cancers, especially those with STK11/LKB1 co-mutation, avoid this response and preferentially activate NF-?B/STAT3 survival signaling has remained unclear. While developing triple combination therapies to inhibit TBK1/MEK signaling and suppress adaptive transcriptional feedback, we made the serendipitous observation that KRAS-LKB1 (KL) mutant lung cancers epigenetically silence STING. Detailed studies in KL cells unveiled a mechanistic connection between enhanced DNMT1 activity and the need to avoid detection of mitochondrial DNA, which accumulates due to damaged mitochondria. Re-activating STING expression in KL cells results in cellular cytotoxicity and enhanced immunogenicity, especially when combined with STING agonism. Thus, instead of inhibiting multiple signaling pathways downstream KRAS, these studies uncover a straightforward vulnerability that could be more readily co-opted therapeutically. The broad, long term objective of this proposal is therefore to characterize the epigenetic mechanism of STING silencing and to co-opt this state into a tumor vulnerability. Forcing cells to deal with the consequences of STING re-expression, while driving its activity via DNA damage or direct STING agonism, has important therapeutic potential for this major subset of KRAS-driven lung cancer. Given the unclear therapeutic window of targeting three or more KRAS downstream pathways, which is required for long-term durable response in animal models, this simpler strategy, which can also re-engage anti-tumor immunity, has significant potential. Moreover, in addition to the in vivo studies we propose, our novel model system using patient-derived xenograft and direct patient-derived organotypic tumor spheroids (XDOTS and PDOTS) provides rapid validation in actual explanted tumors.
Specific aims are to: 1) Optimize strategies to reverse epigenetic silencing of STING in KL tumors, 2) Develop combination therapy strategies with specific agents that promote mitotic slippage, and 3) Utilize immune-competent models to explore the direct role of STING priming on adaptive T cell responses. Through these complementary studies, the goal is to rewire the cytokine circuitry of KRAS-driven lung cancer to engage this cytotoxic anti-viral signaling machinery and ultimately to overcome intrinsic resistance to immune checkpoint inhibitor therapy.

Public Health Relevance

The goal of the proposed research is to understand how certain KRAS-driven lung cancers silence STING and to develop pre-clinical therapeutic strategies that force them to deal with re-activation of this critical DNA sensor. By inducing this antiviral protein and boosting its activity, KRAS-LKB1 mutant lung cancers are forced to deal with consequences, which not only promotes cellular cytotoxicity but also enhanced immunogenicity. These studies build upon our initial efforts to target TBK1 and provide a more straightforward approach to rewire its activity in lung cancer and ultimately have a significant impact on public health.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
2R01CA190394-06A1
Application #
10051766
Study Section
Developmental Therapeutics Study Section (DT)
Program Officer
Venkatachalam, Sundaresan
Project Start
2015-04-01
Project End
2025-07-31
Budget Start
2020-06-01
Budget End
2021-07-31
Support Year
6
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Dana-Farber Cancer Institute
Department
Type
DUNS #
076580745
City
Boston
State
MA
Country
United States
Zip Code
02215
Aref, Amir R; Campisi, Marco; Ivanova, Elena et al. (2018) 3D microfluidic ex vivo culture of organotypic tumor spheroids to model immune checkpoint blockade. Lab Chip 18:3129-3143
Kitajima, Shunsuke; Ivanova, Elena; Guo, Sujuan et al. (2018) Suppression of STING Associated with LKB1 Loss in KRAS-Driven Lung Cancer. Cancer Discov :
Terai, Hideki; Kitajima, Shunsuke; Potter, Danielle S et al. (2018) ER Stress Signaling Promotes the Survival of Cancer ""Persister Cells"" Tolerant to EGFR Tyrosine Kinase Inhibitors. Cancer Res 78:1044-1057
Kitajima, Shunsuke; Barbie, David A (2018) RASA1/NF1-Mutant Lung Cancer: Racing to the Clinic? Clin Cancer Res 24:1243-1245
CaƱadas, Israel; Thummalapalli, Rohit; Kim, Jong Wook et al. (2018) Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses. Nat Med 24:1143-1150
Jenkins, Russell W; Aref, Amir R; Lizotte, Patrick H et al. (2018) Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids. Cancer Discov 8:196-215
Yang, Shenghong; Imamura, Yu; Jenkins, Russell W et al. (2016) Autophagy Inhibition Dysregulates TBK1 Signaling and Promotes Pancreatic Inflammation. Cancer Immunol Res 4:520-30
Koyama, Shohei; Akbay, Esra A; Li, Yvonne Y et al. (2016) STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment. Cancer Res 76:999-1008
Kitajima, Shunsuke; Thummalapalli, Rohit; Barbie, David A (2016) Inflammation as a driver and vulnerability of KRAS mediated oncogenesis. Semin Cell Dev Biol 58:127-35
Van Allen, Eliezer M; Golay, Hadrien G; Liu, Yan et al. (2015) Long-term Benefit of PD-L1 Blockade in Lung Cancer Associated with JAK3 Activation. Cancer Immunol Res 3:855-63

Showing the most recent 10 out of 11 publications