Recent advances have credentialed immunotherapy as a potent anti-cancer treatment modality. However, with exception of high-risk neuroblastoma (NB), immunotherapy has not yet been shown to improve outcomes for children with solid tumors. For patients with NB undergoing myeloablative therapy and autologous hematopoietic stem cell transplantation, subsequent treatment with dinutuximab, which targets disialoganglioside (GD2) on NB cells, improves 2-year event-free survival from 465% to 665%. However, 40% of patients relapse during or after receiving dinutuximab; regimen-related toxicity is high; and 15% of patients have early disease progression and do not receive this therapy. Other immunotherapies include adoptive cell therapy with chimeric antigen receptor (CAR) modified T cells and checkpoint inhibition with monoclonal antibodies (mAbs). CAR T cells have potent activity against pediatric acute lymphoblastic leukemia but have not yet demonstrated activity against solid tumors. Checkpoint inhibition alone (e.g., anti- PD-1 mAb) may not have significant anti-NB activity due to its efficacy correlating with the frequent tumor cell mutations and to the paucity of mutations in NB. Our strategy focuses on cell and mAb based immunotherapies for NB that does not require high-level inherent immunogenicity and builds upon recent advances in our understanding of the tumor cell:immune system interface. We hypothesize that therapeutic gains will be greatest if strategies both enhance cell intrinsic functions of CAR T cells and activated NK (aNK) cells, and overcome cell extrinsic immunosuppression in the tumor microenvironment (TME).
Our Specific Aims are to enhance the potency and durability of CAR T cells and aNK cells and to develop regimens that combine enhanced cell therapies with modulation of the immunosuppressive TME. Our Research Strategy/Approach is 1) to enhance the potency and durability of GD2-CARs using targeted mutagenesis of the scFv to prevent antigen-independent tonic signaling and exhaustion originating from the scFv; to test a novel B7-H3-CAR with demonstrated activity; and to add PD-1 blockade to enhance potency and limit exhaustion; 2) to increase trafficking, persistence and potency of aNK cells by combining them with anti-GD2 and anti-B7-H3 mAbs and with a superagonist IL-15; and 3) to overcome immune suppression in the TME with an anti-CD105 mAb to suppress/eliminate mesenchymal stroma and endothelial cells, with a CSF1R inhibitor to eliminate suppressive MDSC and TAMs (collaboration with Project 4), with a TGFBR1 inhibitor to prevent suppression by TGF?1, and with a FAK/ALK inhibitor to overcome FAK-based induction of T cell suppression and exhaustion (collaboration with Project 1). In summary, novel approaches to enhance the potency and durability of adoptive T cell and NK cell therapies will be developed for translation into clinical trials conducted by Core B (NANT consortium) for patients with high-risk NB.

Public Health Relevance

Recent clinical advances have credentialed immunotherapy as a potent tool in the battle against cancer, and immunotherapy with an anti-neuroblastoma antibody has improved the outcome for children with high-risk neuroblastoma. This project will identify new strategies to further improve potency and durability of anti- neuroblastoma immunotherapy with T cells that are genetically engineered to express neuroblastoma-targeting chimeric antigen receptors and with activated natural killer cells that are combined with neuroblastoma- targeting antibodies. The efficacy of these strategies will be maximized by combining them with regimens that overcome immunosuppression in the tumor microenvironment in collaboration with other Program investigators.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA217959-02
Application #
9567119
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2018-09-01
Budget End
2019-08-31
Support Year
2
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Children's Hospital of Los Angeles
Department
Type
DUNS #
052277936
City
Los Angeles
State
CA
Country
United States
Zip Code
90027
Pinto, Navin; DuBois, Steven G; Marachelian, Araz et al. (2018) Phase I study of vorinostat in combination with isotretinoin in patients with refractory/recurrent neuroblastoma: A new approaches to Neuroblastoma Therapy (NANT) trial. Pediatr Blood Cancer 65:e27023
Iniguez, Amanda Balboni; Alexe, Gabriela; Wang, Emily Jue et al. (2018) Resistance to Epigenetic-Targeted Therapy Engenders Tumor Cell Vulnerabilities Associated with Enhancer Remodeling. Cancer Cell 34:922-938.e7
An, Zhenyi; Aksoy, Ozlem; Zheng, Tina et al. (2018) Epidermal growth factor receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies. Oncogene 37:1561-1575
Villablanca, Judith G; Ji, Lingyun; Shapira-Lewinson, Adi et al. (2018) Predictors of response, progression-free survival, and overall survival using NANT Response Criteria (v1.0) in relapsed and refractory high-risk neuroblastoma. Pediatr Blood Cancer 65:e26940
Tolbert, Vanessa P; Matthay, Katherine K (2018) Neuroblastoma: clinical and biological approach to risk stratification and treatment. Cell Tissue Res 372:195-209
Rajbhandari, Presha; Lopez, Gonzalo; Capdevila, Claudia et al. (2018) Cross-Cohort Analysis Identifies a TEAD4-MYCN Positive Feedback Loop as the Core Regulatory Element of High-Risk Neuroblastoma. Cancer Discov 8:582-599
Fan, Qi Wen; Nicolaides, Theodore P; Weiss, William A (2018) Inhibiting 4EBP1 in Glioblastoma. Clin Cancer Res 24:14-21
Hadjidaniel, Michael D; Muthugounder, Sakunthala; Hung, Long T et al. (2017) Tumor-associated macrophages promote neuroblastoma via STAT3 phosphorylation and up-regulation of c-MYC. Oncotarget 8:91516-91529
Lifshitz, Veronica; Priceman, Saul J; Li, Wenzhao et al. (2017) Sphingosine-1-Phosphate Receptor-1 Promotes Environment-Mediated and Acquired Chemoresistance. Mol Cancer Ther 16:2516-2527
Borriello, Lucia; Nakata, Rie; Sheard, Michael A et al. (2017) Cancer-Associated Fibroblasts Share Characteristics and Protumorigenic Activity with Mesenchymal Stromal Cells. Cancer Res 77:5142-5157