The goal of this project is to develop novel, synthetic lethal, replication stress related, approaches for SCLC patients based on their tumor?s molecular profiles. There are two major unmet needs for SCLC therapy: first, the majority of patients will experience primary or acquired resistance to clinically available chemotherapy and immunotherapy; and second, despite new data supporting molecularly and biologically distinct subsets of SCLC, there are no established approaches for biomarker-driven personalized treatments (such as is standard for targetable oncogenic drivers in NSCLC). Many of the currently ?undruggable? oncogenic changes that are common in SCLC promote tumorigenesis through replication stress (RS) and genomic instability. These ?hallmarks of cancer? lead to high tumor mutational burden and chromosomal aberrations. SCLCs have a continuous high degree of RS due to the universal loss of p53 and RB1 function, and frequent activation of oncogenes such as MYC. In the presence of RS, cancer cells depend on RS responses (RSR), which are a branch of DNA damage repair responses (DDR), to resolve DNA damage and stalled replication forks. Several RSR inhibitors are in clinical development, and we have preliminary findings in preclinical models indicating that SCLC lines, xenografts, and genetically engineered mouse models (GEMMs) are sensitive to agents that target RSR or that increase RS or genomic instability beyond tolerable levels. The latter group includes aurora kinase inhibitors (which induce mitotic catastrophe in the setting of high RS levels, as well as directly inhibiting DNA repair and decreasing replication fork stability) and 6-thio-dG (distinct from the well-known drug 6 thioguanine) which is incorporated by telomerase positive cells, leading to telomere disruption, and increased RS. Furthermore, we have recently found that therapeutic targeting of RS (e.g., by inhibitors of Chk1 or PARP) increases cytoplasmic DNA, resulting in activation of the innate immune response (via the Stimulator of Interferon Genes [STING] pathway), immune-mediated cytotoxicity, and enhanced response to immune checkpoint blockade. We hypothesize that: 1. RS targeting (through Aurora Kinase inhibition or telomere disruption) will lead to synthetic lethality in molecularly-defined subsets of SCLC (Aim 1); and 2. that RS targeting will enhance response to immune checkpoint blockade in immunotherapy-refractory subsets of SCLC via activation of the innate immune system (Aims 2 and 3). Specifically, we expect that RS-targeted therapies will lead to replication catastrophe and cell death in oncogene-driven lung cancers and enhance responses to immune checkpoint blockade (e.g., anti-PD-L1). We will test our hypotheses in an extensive panel of molecularly characterized SPORE lung cancer cell lines, patient derived xenografts (PDXs, tumor and circulating tumor cell (CTC)-derived), GEM-derived animal models (Aims 1 and 2), and in clinical specimens from an investigator-initiated Phase 2 clinical trial (Aim 3) of Aurora Kinase inhibition combined with checkpoint inhibitor immunotherapy in extensive- stage SCLC.

Public Health Relevance

Progress is slow in treating advanced small cell lung cancers (SCLC), and there remains an urgent need to provide patients with long-term durable responses. Approaches to develop SCLC targeted therapy, improve responses to immunotherapy, and to more effectively treat primary chemotherapy relapsed and refractory SCLC are acutely needed. Our Project addresses all of these aspects by testing targeted therapy induced replication stress (RS) as a potential SCLC therapeutic vulnerability which also would increase innate immune responses against SCLC, and lead to improved responses against immune checkpoint blockade immunotherapy. This new SCLC RS vulnerability is studied through a series of preclinical translational experiments and an investigator- initiated Phase 2 clinical trial.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
2P50CA070907-21A1
Application #
10023868
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
Ng, Patrick Kwok-Shing; Li, Jun; Jeong, Kang Jin et al. (2018) Systematic Functional Annotation of Somatic Mutations in Cancer. Cancer Cell 33:450-462.e10
Cascone, Tina; Gold, Kathryn A; Swisher, Stephen G et al. (2018) Induction Cisplatin Docetaxel Followed by Surgery and Erlotinib in Non-Small Cell Lung Cancer. Ann Thorac Surg 105:418-424
Kim, Wanil; Shay, Jerry W (2018) Long-range telomere regulation of gene expression: Telomere looping and telomere position effect over long distances (TPE-OLD). Differentiation 99:1-9
Wang, Min; Abrams, Zachary B; Kornblau, Steven M et al. (2018) Thresher: determining the number of clusters while removing outliers. BMC Bioinformatics 19:9
Sinicropi-Yao, Sara L; Amann, Joseph M; Lopez, David Lopez Y et al. (2018) Co-Expression Analysis Reveals Mechanisms Underlying the Varied Roles of NOTCH1 in NSCLC. J Thorac Oncol :
Le, Xiuning; Puri, Sonam; Negrao, Marcelo V et al. (2018) Landscape of EGFR-Dependent and -Independent Resistance Mechanisms to Osimertinib and Continuation Therapy Beyond Progression in EGFR-Mutant NSCLC. Clin Cancer Res 24:6195-6203
Wang, Shidan; Chen, Alyssa; Yang, Lin et al. (2018) Comprehensive analysis of lung cancer pathology images to discover tumor shape and boundary features that predict survival outcome. Sci Rep 8:10393
Gomez, Daniel Richard; Byers, Lauren Averett; Nilsson, Monique et al. (2018) Integrative proteomic and transcriptomic analysis provides evidence for TrkB (NTRK2) as a therapeutic target in combination with tyrosine kinase inhibitors for non-small cell lung cancer. Oncotarget 9:14268-14284
Parra, Edwin R; Villalobos, Pamela; Mino, Barbara et al. (2018) Comparison of Different Antibody Clones for Immunohistochemistry Detection of Programmed Cell Death Ligand 1 (PD-L1) on Non-Small Cell Lung Carcinoma. Appl Immunohistochem Mol Morphol 26:83-93
Yamauchi, Mitsuo; Barker, Thomas H; Gibbons, Don L et al. (2018) The fibrotic tumor stroma. J Clin Invest 128:16-25

Showing the most recent 10 out of 1059 publications