Using a novel membrane-anchored IL-12 T cell therapy that binds to tumor cell surface vimentin (attIL12), Hu et al. investigated how to reshape ECM-rich tumors, such as osteosarcoma. In an in vitro model of patient-derived osteosarcoma cells, gene expression studies showed that attIL12-armed T cells disrupted the ECM and destroyed cancer-associated fibroblasts (CAFs). Mechanistically, attIL12 treatment increased IFNγ production in the TME, which led to upregulation of FAS-mediated CAF apoptosis. CAF apoptosis led to TGFβ downregulation and impaired collagen and fibronectin production, increasing the susceptibility of ECM-rich tumors to T cell-mediated killing.

Contributed by Katherine Turner

BACKGROUND: The extracellular matrix (ECM) and cancer-associated fibroblasts (CAFs) play major roles in tumor progression, metastasis, and the poor response of many solid tumors to immunotherapy. CAF-targeted chimeric antigen receptor-T cell therapy cannot infiltrate ECM-rich tumors such as osteosarcoma. METHOD: In this study, we used RNA sequencing to assess whether the recently invented membrane-anchored and tumor-targeted IL-12-armed (attIL12) T cells, which bind cell-surface vimentin (CSV) on tumor cells, could destroy CAFs to disrupt the ECM. We established an in vitro model of the interaction between osteosarcoma CAFs and attIL12-T cells to uncover the underlying mechanism by which attIL12-T cells penetrate stroma-enriched osteosarcoma tumors. RESULTS: RNA sequencing demonstrated that attIL12-T cell treatment altered ECM-related gene expression. Immunohistochemistry staining revealed disruption or elimination of high-density CAFs and ECM in osteosarcoma xenograft tumors following attIL12-T cell treatment, and CAF/ECM density was inversely correlated with T-cell infiltration. Other IL12-armed T cells, such as wild-type IL-12-targeted or tumor-targeted IL-12-T cells, did not disrupt the ECM because this effect depended on the engagement between CSV on the tumor cell and its ligand on the attIL12-T cells. Mechanistic studies found that attIL12-T cell treatment elevated IFN_ production on interacting with CSV(+) tumor cells, suppressing transforming growth factor beta secretion and in turn upregulating FAS-mediated CAF apoptosis. CAF destruction reshaped the tumor stroma to favor T-cell infiltration and tumor inhibition. CONCLUSIONS: This study unveiled a novel therapy-attIL12-T cells-for targeting CAFs/ECM. These findings are highly relevant to humans because CAFs are abundant in human osteosarcoma.

Author Info: (1) Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (2) Department of Pathology, The University of Texas MD Anderson Canc

Author Info: (1) Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (2) Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. Department of Genomic Medicine, The Universiy of Texas MD Anderson Cancer Center, Houston, Texas, USA. (3) Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (4) Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (5) Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (6) Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (7) Department of Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (8) Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (9) Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (10) Department of Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (11) Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. (12) Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA sli4@mdanderson.org.