Poster Session A   |   11:45am Expo - Hall A & C   |   Poster ID #235

Production and Functional Characterization of HLA-G Targeting Chimeric Antigen Receptor (CAR) expressing Natural Killer (NK) Cells for the Treatment of Hematological and Solid malignancies.

Program:
Academic Research
Category:
Immunology
FDA Status:
Not Applicable
CPRIT Grant:
Cancer Site(s):
Pancreas, Brain and Nervous System, Leukemias
Authors:
Kiran Kundu
The University of Texas M.D. Anderson Cancer Center
Rafet Basar
The University of Texas M.D. Anderson Cancer Center
Sunil Acharaya
The University of Texas M.D. Anderson Cancer Center
Nadima Uprety
The University of Texas M.D. Anderson Cancer Center
Katy Rezvani
The University of Texas M.D. Anderson Cancer Center

Introduction

Chimeric antigen receptor (CAR) expressing natural killer (NK) cell-based immunotherapy is offering a paradigm shift in personalized cancer medicine as they can be utilized as universal cellā€based therapy without requiring human leukocyte antigen (HLA) matching like T cells. Though NK cell based clinical trials are ongoing, identification of novel targets is necessary to achieve better efficacy. Human leukocyte antigen G (HLA-G) is a central protein that plays a critical role in providing immune tolerance to the fetus in pregnant women. As a class I major histocompatibility complex (MHC) protein, HLA-G suppresses the function of NK cells and a subset of T cells via engagement with inhibitory receptors such as leukocyte Ig-like receptor subfamily B member 1 (LILRB1) and member 2 (LILRB2), and the killer immunoglobulin-like receptor 2DL4 (KIR2DL4). Cancer cells aberrantly upregulate HLA-G on their cell surface, thus, inducing immunosuppression within the tumor-microenvironment. This makes HLA-G an attractive cell therapy target. Here, we propose to develop CAR-NK cells targeting HLA-G protein as a novel strategy for cancer immunotherapy, using NK cells from healthy human donor cord blood.

Methods

Flow cytometry-based cell surface staining of HLA-G was done on different cancer cell lines of acute myeloid leukemia (AML), glioblastoma and pancreatic cancer. The mRNA expression analysis of HLA-G isoforms was done on the above-mentioned cancer types using TCGA data set. NK cells were isolated from healthy human donor cord blood via a negative selection kit. After days of expansion with specific feeder layer of cells, transduction was performed with a retroviral construct containing the ScFv sequence derived from a specific human anti-HLA-G monoclonal antibody. In addition, our construct incorporated the gene for interleukin 15 (IL-15), an important cytokine for NK proliferation and persistence in vivo. To determine the role of different costimulatory molecules on HLA-G CAR NK potency, we compared two different signaling domains coming from CD28 and DAP10. CAR-NK cells in vitro functionality was tested by co-culturing with tumor cell lines and further assessing cell death via incucyte and xcelligence instrument. Flow cytometry was performed to measure NK cytotoxic markers e.g., CD107a, TNF-a, IFN-g.

Results

We observed elevated HLA-G surface expression on cell lines from AML, glioblastoma, pancreatic ductal adenocarcinoma, breast cancer and renal cancer. HLA-G isoform mRNA expression analysis from TCGA data also showed differential elevated mRNA expression of HLA-G isoforms on those cancer types.

We obtained an average of 50-70% transduction efficiency and HLA-G CAR expression on cord blood derived NK cells. In short time co-culture experiments, HLA-G CAR NK cells killed OCI-AML3 cells even in the absence of IL-15. HLA-G CAR NK cells also showed cytotoxicity towards GSC20 spheroids and PANC1 cells. When HLA-G CAR NK cells were incubated with plate bound HLA-G protein, they showed strong activation via upregulation of CD107a, TNF-a, IFN-g compared to the non-specific protein CD5. To understand the functionality better, we generated CAR construct with CD28 and DAP10 costimulatory domain. Both costimulatory domains containing constructs also showed robust cytotoxicity against OCI-AML3 cells at short-term as well as long-term tumor rechallenge killing assay.

Conclusion

Our finding suggests that HLA-G is upregulated at protein level in different hematological and solid cancer models and that HLA-G targeting CAR NK cells, generated from human donor cord blood, could be a novel off-the-shelf cellular therapy approach for different malignancies.

Tumor xenograft experiments to study the in vivo safety and efficacy of HLA-G targeting CAR NK cells are under way.