Neuroblastoma (NB) remains one of the most difficult pediatric cancers to treat, as nearly two-thirds of the patients present with metastasis at the tim of diagnosis. Despite aggressive treatment protocols, `high-risk' NB, stage 4 tumors in children older than 18 months of age and/or MYCN amplification, have dismal overall survival of only 50%. There is a significant gap in the comprehensive understanding of the tumor biology for tumor refractoriness and disease relapse, and therefore, elucidation of signaling mechanisms responsible for the aggressive tumor behavior would be highly significant for development of novel targeted clinical therapies. Members of the G-protein coupled receptor (GPCR) superfamily represent the hub of drug development activities and accounts for 40% of all targeted therapies. Our laboratory has previously shown that targeting one such GPCR, gastrin-releasing peptide receptor (GRP-R) and its ligand GRP inhibited NB tumorigenicity via regulation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, a key cell survival pathway. During the previous funding period (see Progress Report), we reported that the GRP-R signaling regulates NB metastasis, specifically through activation of AKT2, and downregulation of PTEN, an endogenous negative regulator of AKT. The exact role of the GRP-R signaling and specific AKT isoform, AKT2, in the multi-step NB progression from tissue invasion to distant organ metastasis is yet to be elucidated. Targeting PI3K/AKT2, as well as mTOR, downstream of AKT, axis may be a potential strategy as this pathway regulates tumor vasculature and microenvironment (TME). In this competitive renewal application, we plan to determine the exact mechanisms by which GRP-R/AKT2 axis regulates the formation and maintenance of metastasis-initiating cells and their dissemination to distant organs and facilitate establishment of metastatic lesions. Hence, the central hypothesis of this proposal is that GRP/GRP-R signaling via AKT2 regulates resistance to conventional therapies and formation of metastatic foci, thereby, implicating a critical role for this axis in NB refractoriness and disease relapse. o examine this hypothesis, we propose the following three Specific Aims: 1) Determine the role of GRP-R/AKT2 signaling in inducing resistance to conventional therapies and tumor progression. 2) Determine the role of AKT2 in mediating the oncogenic effects of GRP/GRP-R in refractory and/or metastatic NB. 3) Determine the preclinical efficacy of targeting GRP-R/AKT2 in refractory and/or metastatic NB. Successful completion of our Aims will yield significant new knowledge regarding the mechanisms of GRP/GRP-R mediated NB refractoriness and disease relapse, including the role of cancer stem cell in metastasis as well as the extravasation of tumor cells and their interactions with TME.
The aims proposed have direct translational relevance as they address an important molecular mechanism of resistance to conventional therapies and the TME. Finally, preclinical study will further provide important information on the safety and efficacy of novel therapeutics against virulent `high-risk' NB.

Public Health Relevance

Despite recent advances in multi-modality aggressive treatment protocols, infants and children with `high-risk' category of advanced-stage neuroblastoma with a high incidence of metastasis still have a staggering overall disease-free survival of less than 50%. Our project is clinically significant and has direct translational relevance because it will generate critical mechanistic insights to tumor refractoriness and relapse, which could lead to a breakthrough in the treatment of this devastating disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
2R01DK061470-11
Application #
8888937
Study Section
Bioengineering, Technology and Surgical Sciences Study Section (BTSS)
Program Officer
Serrano, Jose
Project Start
2002-04-01
Project End
2019-03-31
Budget Start
2015-04-01
Budget End
2016-03-31
Support Year
11
Fiscal Year
2015
Total Cost
Indirect Cost
Name
Vanderbilt University Medical Center
Department
Surgery
Type
Schools of Medicine
DUNS #
004413456
City
Nashville
State
TN
Country
United States
Zip Code
37240
Rellinger, Eric J; Padmanabhan, Chandrasekhar; Qiao, Jingbo et al. (2017) ML327 induces apoptosis and sensitizes Ewing sarcoma cells to TNF-related apoptosis-inducing ligand. Biochem Biophys Res Commun 491:463-468
Padmanabhan, Chandrasekhar; Rellinger, Eric J; Zhu, Jing et al. (2017) cFLIP critically modulates apoptotic resistance in epithelial-to-mesenchymal transition. Oncotarget 8:101072-101086
Rellinger, Eric J; Craig, Brian T; Alvarez, Alexandra L et al. (2017) FX11 inhibits aerobic glycolysis and growth of neuroblastoma cells. Surgery 161:747-752
Paul, Pritha; Rellinger, Eric J; Qiao, Jingbo et al. (2017) Elevated TIMP-1 expression is associated with a prometastatic phenotype, disease relapse, and poor survival in neuroblastoma. Oncotarget 8:82609-82620
Rellinger, Eric J; Padmanabhan, Chandrasekhar; Qiao, Jingbo et al. (2017) Isoxazole compound ML327 blocks MYC expression and tumor formation in neuroblastoma. Oncotarget 8:91040-91051
Craig, Brian T; Rellinger, Eric J; Alvarez, Alexandra L et al. (2016) Induced differentiation inhibits sphere formation in neuroblastoma. Biochem Biophys Res Commun 477:255-9
Qiao, Jingbo; Grabowska, Magdalena M; Forestier-Roman, Ingrid S et al. (2016) Activation of GRP/GRP-R signaling contributes to castration-resistant prostate cancer progression. Oncotarget 7:61955-61969
Mobley, Bret C; Kwon, Minjae; Kraemer, Bradley R et al. (2015) Expression of MYCN in Multipotent Sympathoadrenal Progenitors Induces Proliferation and Neural Differentiation, but Is Not Sufficient for Tumorigenesis. PLoS One 10:e0133897
Zhu, Yueming; Paul, Pritha; Lee, Sora et al. (2015) Antioxidant inhibition of steady-state reactive oxygen species and cell growth in neuroblastoma. Surgery 158:827-36
Rellinger, Eric J; Romain, Carmelle; Choi, SunPhil et al. (2015) Silencing gastrin-releasing peptide receptor suppresses key regulators of aerobic glycolysis in neuroblastoma cells. Pediatr Blood Cancer 62:581-6

Showing the most recent 10 out of 56 publications