Lung cancer is the leading cause of cancer deaths worldwide, with 5-year survival rates of <15%, and this is largely due to the lack of effective treatments for advanced disease. Activating mutations of the K-ras gene are found in more than 30% of lung adenocarcinoma, indicating that mutant K- ras is a driver of lung cancer and an important target for therapeutic intervention. However, previous attempts to directly target mutant K-ras have met with little success. Thus, alternative approaches to target activated K-Ras signaling are warranted. Our long-term goals are to identify effectors that are required for mutant K-Ras-driven tumorigenesis and to develop therapeutic approaches to targets these molecules and pathways. Our Preliminary Studies, along with recent findings from others, suggest that Notch signaling pathways are potentially important therapeutic targets in non-small cell lung cancer (NSCLC). The Notch family consists of four receptors in mammals (Notch1-4) and current approaches to Notch inhibition involve the use of ?-secretase inhibitors (GSIs), which inhibit the activation of all four receptors. Unfortunately, the use of GSIs in patients is limited due to severe intestinal toxicities that are a result of global Notch inhibition. In additin, recent studies indicate that the different Notch receptors have distinct and non-overlapping functions in tumorigenesis and that these functions are cell type and timing-dependent. These finding suggest that anti-cancer therapies targeting Notch receptors and their downstream signaling pathways need to be tailored to individual receptors. Given previous findings suggesting Notch1 is required for K-ras-induced tumorigenesis, we tested whether Notch1 is necessary for the development of mutant K-ras-driven NSCLC in vivo, by crossing the conditionally inducible K-rasG12D knock-in mouse model with a Notch1 conditional knockout mouse. In these mice, Cre-mediated recombination simultaneously activates the K-rasG12D allele and inactivates Notch1. We find that loss of Notch1 significantly reduced tumor initiation and overall tumor burden, indicating that Notch1 is required for K-ras-induced lung tumorigenesis. Moreover, our studies have shown that Notch1 contributes to K-ras-driven NSCLC by suppressing p53-mediated apoptosis. Importantly, these results identify the molecular basis for the documented correlation between Notch1 activity and poor prognosis in NSCLC patients having wild type p53, which represent 50% of all NSCLC cases. Based on our findings, we hypothesize that Notch1 is required for lung tumorigenesis via its ability to suppress p53, and that targeting Notch1 and/or its downstream effectors will inhibit the development and/or maintenance of NSCLC. The proposed studies will test this hypothesis and define the mechanisms underlying the requirement for Notch1 in K-Ras driven NSCLC. In addition, we will test the role of Notch1 in NSCLC driven by oncogenic events other than K-ras mutation, to determine if Notch1- directed therapies will provide benefit to a broad spectrum of NSCLC patients.

Public Health Relevance

Lung cancer is the leading cause of cancer deaths worldwide, with 5-year survival rates of <15%, and this is largely due to the lack of effective treatments for advanced disease. Activating mutations of the K-ras gene are found in more than 30% of lung adenocarcinoma, however, previous attempts to directly target mutant K-ras have met with little success. Thus, our long-term goals are to identify effectors that are required for mutant K-Ras-driven tumorigenesis and to develop therapeutic approaches to targets these molecules and pathways. Our Preliminary Studies indicate that Notch1 is necessary for the development of mutant K-ras-driven NSCLC in vivo, by suppression of p53-mediated cell death. Based on our findings, we hypothesize that targeting Notch1 and/or its downstream effectors will inhibit the development and/or maintenance of NSCLC. The proposed studies will test the hypothesis that Notch1 is required for maintenance of established lung tumors in vivo, via knockdown and genetic ablation in transplantable and endogenous models of lung adenocarcinoma, respectively. In addition, candidate-based and unbiased approaches will be used to determine the mechanism by which Notch1 regulates p53 and we will identify and validate critical effectors in this response, which could then be developed as targets towards the treatment of NSCLC. Finally, we will determine if Notch1 functions are required in a broader range of NSCLC, by testing the requirement for Notch1 in the development and maintenance of mutant EGFR-driven NSCLC, as well as in NSCLC induced by a tobacco-specific carcinogen. These studies will determine if Notch1-directed therapies will provide benefit to a broad spectrum of NSCLC patients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA124495-09
Application #
8985664
Study Section
Molecular Oncogenesis Study Section (MONC)
Program Officer
Watson, Joanna M
Project Start
2006-12-01
Project End
2018-12-31
Budget Start
2016-01-01
Budget End
2016-12-31
Support Year
9
Fiscal Year
2016
Total Cost
$388,800
Indirect Cost
$186,300
Name
Scripps Florida
Department
Type
DUNS #
148230662
City
Jupiter
State
FL
Country
United States
Zip Code
33458
Kota, Smitha; Hou, Shurong; Guerrant, William et al. (2018) A novel three-dimensional high-throughput screening approach identifies inducers of a mutant KRAS selective lethal phenotype. Oncogene 37:4372-4384
Moleirinho, Susana; Hoxha, Sany; Mandati, Vinay et al. (2017) Regulation of localization and function of the transcriptional co-activator YAP by angiomotin. Elife 6:
Guerrant, William; Kota, Smitha; Troutman, Scott et al. (2016) YAP Mediates Tumorigenesis in Neurofibromatosis Type 2 by Promoting Cell Survival and Proliferation through a COX-2-EGFR Signaling Axis. Cancer Res 76:3507-19
Troutman, Scott; Moleirinho, Susana; Kota, Smitha et al. (2016) Crizotinib inhibits NF2-associated schwannoma through inhibition of focal adhesion kinase 1. Oncotarget 7:54515-54525
Moleirinho, Susana; Guerrant, William; Kissil, Joseph L (2014) The Angiomotins--from discovery to function. FEBS Lett 588:2693-703
Yi, Chunling; Shen, Zhewei; Stemmer-Rachamimov, Anat et al. (2013) The p130 isoform of angiomotin is required for Yap-mediated hepatic epithelial cell proliferation and tumorigenesis. Sci Signal 6:ra77
Licciulli, Silvia; Maksimoska, Jasna; Zhou, Chun et al. (2013) FRAX597, a small molecule inhibitor of the p21-activated kinases, inhibits tumorigenesis of neurofibromatosis type 2 (NF2)-associated Schwannomas. J Biol Chem 288:29105-14
Avila, Jacqueline L; Kissil, Joseph L (2013) Notch signaling in pancreatic cancer: oncogene or tumor suppressor? Trends Mol Med 19:320-7
Zhou, C; Licciulli, S; Avila, J L et al. (2013) The Rac1 splice form Rac1b promotes K-ras-induced lung tumorigenesis. Oncogene 32:903-9
Licciulli, Silvia; Avila, Jacqueline L; Hanlon, Linda et al. (2013) Notch1 is required for Kras-induced lung adenocarcinoma and controls tumor cell survival via p53. Cancer Res 73:5974-84

Showing the most recent 10 out of 18 publications