NYMC Faculty Publications

Combinatorial Macrophage Induced Innate Immunotherapy Against Ewing Sarcoma: Turning "Two Keys" Simultaneously

Authors

Wen Luo, Department of Pediatrics, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA. Wen_Luo@nymc.edu.
Hai Hoang, Department of Pediatrics, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA.
Katherine E. Miller, Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
Hongwen Zhu, Department of Pediatrics, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA.
Serena Xu, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA.
Xiaokui Mo, Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University, Columbus, OH, USA.
Elizabeth A. Garfinkle, Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
Heather Costello, Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
Saranga Wijeratne, Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
Wiebke Chemnitz, Department of Medicine, New York Medical College, Valhalla, NY, USA.
Ronan Gandhi, University of Chicago, Chicago, IL, USA.
Yanling Liao, Department of Pediatrics, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA.
Janet Ayello, Department of Pediatrics, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA.
Aliza Gardenswartz, Department of Pediatrics, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA.
Jeremy M. Rosenblum, Department of Pediatrics, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA.
Kevin A. Cassady, Center for Childhood Cancer Research, Nationwide Children's Hospital, Columbus, OH, USA.
Elaine R. Mardis, Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
Dean A. Lee, Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
Timothy P. Cripe, Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
Mitchell S. Cairo, Department of Pediatrics, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA. Mitchell_Cairo@nymc.edu.

Author Type(s)

Faculty

DOI

10.1186/s13046-024-03093-w

Journal Title

Journal of Experimental & Clinical Cancer Research

First Page

193

Document Type

Article

Publication Date

7-11-2024

Department

Pediatrics

Abstract

BACKGROUND: Macrophages play important roles in phagocytosing tumor cells. However, tumors escape macrophage phagocytosis in part through the expression of anti-phagocytic signals, most commonly CD47. In Ewing sarcoma (ES), we found that tumor cells utilize dual mechanisms to evade macrophage clearance by simultaneously over-expressing CD47 and down-regulating cell surface calreticulin (csCRT), the pro-phagocytic signal. Here, we investigate the combination of a CD47 blockade (magrolimab, MAG) to inhibit the anti-phagocytic signal and a chemotherapy regimen (doxorubicin, DOX) to enhance the pro-phagocytic signal to induce macrophage phagocytosis of ES cells in vitro and inhibit tumor growth and metastasis in vivo. METHODS: Macrophages were derived from human peripheral blood monocytes by granulocyte-macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF). Flow cytometry- and microscopy-based in-vitro phagocytosis assays were performed to evaluate macrophage phagocytosis of ES cells. Annexin-V assay was performed to evaluate apoptosis. CD47 was knocked out by CRISPR/Cas9 approach. ES cell-based and patient-derived-xenograft (PDX)-based mouse models were utilized to assess the effects of MAG and/or DOX on ES tumor development and animal survival. RNA-Seq combined with CIBERSORTx analysis was utilized to identify changes in tumor cell transcriptome and tumor infiltrating immune cell profiling in MAG and/or DOX treated xenograft tumors. RESULTS: We found that MAG significantly increased macrophage phagocytosis of ES cells in vitro (p < 0.01) and had significant effect on reducing tumor burden (p < 0.01) and increasing survival in NSG mouse model (p < 0.001). The csCRT level on ES cells was significantly enhanced by DOX in a dose- and time-dependent manner (p < 0.01). Importantly, DOX combined with MAG significantly enhanced macrophage phagocytosis of ES cells in vitro (p < 0.01) and significantly decreased tumor burden (p < 0.01) and lung metastasis (p < 0.0001) and extended animal survival in vivo in two different mouse models of ES (p < 0.0001). Furthermore, we identified CD38, CD209, CD163 and CD206 as potential markers for ES-phagocytic macrophages. Moreover, we found increased M2 macrophage infiltration and decreased expression of Cd209 in the tumor microenvironment of MAG and DOX combinatorial therapy treated tumors. CONCLUSIONS: By turning "two keys" simultaneously to reactivate macrophage phagocytic activity, our data demonstrated an effective and highly translatable alternative therapeutic approach utilizing innate (tumor associated macrophages) immunotherapy against high-risk metastatic ES.

This document is currently not available here.

Share

COinS