High-risk neuroblastoma (NB) remains a major challenge in pediatric oncology accounting for 15% of all pediatric cancer mortality with an overall 5-year survival of less than 50% for this aggressive embryonal malignancy. As with 90% of all solid tumors, the primary cause of death for NB is relapsed drug-resistant disease. Significant improvements in cure rates for NB will require novel biologically specific approaches since chemotherapy at maximal tolerated doses still fails in over half of theses cases. Models of cancer stem cells (CSCs) now suggest that treatment failure and relapse is often driven by a small drug-resistant subpopulation of self-renewing tumor initiating cells. Our therapeutic goal is to combine anti-CSC therapies with current chemotherapy approaches to both improve initial drug responses and prevent recurrent metastatic disease. Preliminary Data: We have recently demonstrated the isolation and characterization of a highly tumorigenic subpopulation based on the expression of CD114 (the surface receptor for granulocyte colony-stimulating factor (G-CSF)), which fulfills the major phenotypic requirements for tumor initiating CSCs. This tumor subpopulation is highly enriched (up to 10-fold) after chemotherapy in NB tumors resected after 3 cycles of chemotherapy and has gene expression, microRNA expression, and epigenetic profiles which recapitulate those of iPSCs (induced pluripotent stem cells) and ESCs (embryonic stem cells). STAT3 activation through phosphorylation (pSTAT3) is the canonical downstream effector of G-CSF ligand binding to its receptor (CD114). pSTAT3 has a central role in regulating the maintenance of normal and malignant stem cell populations in part through transcriptional regulation of microRNAs involved in reprogramming and epithelial-mesenchymal transitions (EMT). We demonstrate within the CD114+ CSC-like cells, G-CSF dependent pSTAT3 activation results in increased expression of STAT3 target microRNAs. We also demonstrate that the CSFR3 gene (encoding CD114) is a direct transcriptional target of pSTAT3, suggesting a feedback loop for maintenance of expression of this receptor. We therefore hypothesize the following: a) the G-CSF/STAT3 signaling axis promotes the maintenance, drug resistance and metastatic potential of CD114+ NB cells b) Targeting CD114+ cells, either directly or via G- CSF/STAT3 pathway inhibition will limit tumor proliferation and the development of metastasis. C) Concomitant targeting of both the CD114+ and CD114- subpopulations in neuroblastoma should significantly augment chemotherapies by preventing regeneration of CD114- cells from the CD114+ precursors.
In Specific Aims 1 and 2, we will block the G-CSF/STAT3 axis in CD114+ NB cells using by several independent approaches (anti-receptor antibody, JAK/STAT small molecule inhibitors) to determine the role of G-CSF signaling in CD114+ driven NB tumorigenesis and metastasis.
In Aim 3 we will determine how STAT3 target microRNAs contribute to the stemness phenotype of CD114+ cells.

Public Health Relevance

Cancer stem cells (CSCs) may drive disease relapse which accounts for the majority of deaths in neuroblastoma (NB). We will determine how STAT3 signaling, downstream of the G-CSF growth factor, contributes to the tumorigenicity and maintenance of a novel G-CSF receptor + CSC-like NB subpopulation.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA174808-06
Application #
9487924
Study Section
Molecular Oncogenesis Study Section (MONC)
Program Officer
Snyderwine, Elizabeth G
Project Start
2014-06-09
Project End
2019-05-31
Budget Start
2018-06-01
Budget End
2019-05-31
Support Year
6
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of Massachusetts Medical School Worcester
Department
Pediatrics
Type
Schools of Medicine
DUNS #
603847393
City
Worcester
State
MA
Country
United States
Zip Code
Tomolonis, Julie A; Agarwal, Saurabh; Shohet, Jason M (2018) Neuroblastoma pathogenesis: deregulation of embryonic neural crest development. Cell Tissue Res 372:245-262
Agarwal, Saurabh; Milazzo, Giorgio; Rajapakshe, Kimal et al. (2018) MYCN acts as a direct co-regulator of p53 in MYCN amplified neuroblastoma. Oncotarget 9:20323-20338
Sun, Wenjing; Rojas, Yesenia; Wang, Hao et al. (2017) EWS-FLI1 and RNA helicase A interaction inhibitor YK-4-279 inhibits growth of neuroblastoma. Oncotarget 8:94780-94792
Van Goethem, Alan; Yigit, Nurten; Moreno-Smith, Myrthala et al. (2017) Dual targeting of MDM2 and BCL2 as a therapeutic strategy in neuroblastoma. Oncotarget 8:57047-57057
Ghaghada, Ketan B; Starosolski, Zbigniew A; Lakoma, Anna et al. (2016) Heterogeneous Uptake of Nanoparticles in Mouse Models of Pediatric High-Risk Neuroblastoma. PLoS One 11:e0165877
Van Goethem, Alan; Yigit, Nurten; Everaert, Celine et al. (2016) Depletion of tRNA-halves enables effective small RNA sequencing of low-input murine serum samples. Sci Rep 6:37876
Louis, Chrystal U; Shohet, Jason M (2015) Neuroblastoma: molecular pathogenesis and therapy. Annu Rev Med 66:49-63
Agarwal, Saurabh; Lakoma, Anna; Chen, Zaowen et al. (2015) G-CSF Promotes Neuroblastoma Tumorigenicity and Metastasis via STAT3-Dependent Cancer Stem Cell Activation. Cancer Res 75:2566-79
Beckers, Anneleen; Van Peer, Gert; Carter, Daniel R et al. (2015) MYCN-driven regulatory mechanisms controlling LIN28B in neuroblastoma. Cancer Lett 366:123-32
Kim, Eugene S; Agarwal, Saurabh; Shohet, Jason M (2015) G-CSF Is a Cancer Stem Cell-Specific Growth Factor-Response. Cancer Res 75:3992