Fujiwara et al. demonstrated that PLX3397, a potent small molecule CSF-1R inhibitor, reprograms tumor-associated macrophages (TAMs) to exert its antitumor effects. Osteosarcoma and fibrosarcoma cells secreted CSF-1 in conditioned medium, and, in an in vitro TAM model, CSF-1R blockade reduced M2 polarization, proliferation, and chemotaxis in bone marrow-derived macrophages. Systemic administration of PLX3397 in a mouse osteosarcoma model suppressed primary tumor growth and lung metastasis, improved metastasis-free survival, reduced TAMs and Foxp3+ Tregs, and increased the CD8+ T cell infiltration in the TME.

Contributed by Shishir Pant

ABSTRACT: Colony-stimulating factor 1 (CSF-1) is a primary regulator of the survival, proliferation, and differentiation of monocyte/macrophage that sustains the pro-tumorigenic functions of tumor-associated macrophages (TAMs). Considering current advances in understanding the role of the inflammatory tumor microenvironment, targeting the components of the sarcoma microenvironment, such as TAMs, is a viable strategy. Here, we investigated the effect of PLX3397 (pexidartinib) as a potent inhibitor of the CSF-1 receptor (CSF-1R). PLX3397 was recently approved by the Food and Drug Administration (FDA) to treat tenosynovial giant cell tumor and reprogram TAMs whose infiltration correlates with unfavorable prognosis of sarcomas. First, we confirmed by cytokine arrays of tumor-conditioned media (TCM) that cytokines including CSF-1 are secreted from LM8 osteosarcoma cells and NFSa fibrosarcoma cells. The TCM, like CSF-1, stimulated ERK1/2 phosphorylation in bone marrow-derived macrophages (BMDMs), polarized BMDMs toward a M2 (TAM-like) phenotype, and strikingly promoted BMDM chemotaxis. In vitro administration of PLX3397 suppressed pERK1/2 stimulation by CSF-1 or TCM, and reduced M2 polarization, survival, and chemotaxis in BMDMs. Systemic administration of PLX3397 to the osteosarcoma orthotopic xenograft model significantly suppressed the primary tumor growth and lung metastasis, and thus improved metastasis-free survival. PLX3397 treatment concurrently depleted TAMs and FOXP3+ regulatory T cells and, surprisingly, enhanced infiltration of CD8+ T cells into the microenvironments of both primary and metastatic osteosarcoma sites. Our preclinical results show that PLX3397 has strong macrophage- and T cell-modulating effects that may translate into cancer immunotherapy for bone and soft tissue sarcomas.

Author Info: (1) Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine. (2) Orthopedic Surgery, Memorial Sloan Kettering Cancer Center. (3) College of Osteopathic Me

Author Info: (1) Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine. (2) Orthopedic Surgery, Memorial Sloan Kettering Cancer Center. (3) College of Osteopathic Medicine, Tuoro College of Ostopathic Medicine. (4) Orthopaedic Surgery, Keck Hospital of USC. (5) Organic Synthesis Core Laboratory, Memorial Sloan Kettering Cancer Center. (6) Organic Synthesis Core Laboratory, Memorial Sloan Kettering Cancer Center. (7) Orthopaedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center. (8) Department of Orthopedic Surgery, Okayama University Graduate School of Medicine. (9) Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine. (10) Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine. (11) Center for Innovative Clinical Medicine, Okayama University Hospital. (12) Lysosomal Storage Diseases, AVROBIO. (13) Cell Biology Program, Memorial Sloan Kettering Cancer Center. (14) Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine. (15) Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine. (16) Hospital for Special Surgery. (17) Orthopaedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center healeyj@mskcc.org.