Knudson et al. tested M7824, a fusion protein that combines TGFβRII with anti-PD-L1, in murine primary and metastatic models of breast and colon carcinoma. M7824 targeted to the tumor via PD-L1 binding, sequestered TGFβ, and activated NK and CD8+ T cells, leading to reduced tumor growth, increased median overall survival (OS), and memory response in mice with complete tumor rejection. The antitumor efficacy was dependent on NK and CD8+ T cells. M7824 combined with a tumor-associated antigen TWIST1 vaccine further increased OS; unexpectedly, combining vaccine with anti-PD-L1 abrogated the antitumor effect of anti-PD-L1.

Tumors evade host immune surveillance through multiple mechanisms, including the generation of a tumor microenvironment that suppresses immune effector function. Secretion of TGFbeta and upregulation of immune checkpoint programmed cell death ligand-1 (PD-L1) are two main contributors to immune evasion and tumor progression. Here, we examined the efficacy of a first-in-class bifunctional checkpoint inhibitor, the fusion protein M7824, comprising the extracellular domain of human TGFbetaRII (TGFbeta Trap) linked to the C-terminus of human anti-PD-L1 heavy chain (alphaPD-L1). We demonstrate that M7824 reduces plasma TGFbeta1, binds to PD-L1 in the tumor, and decreases TGFbeta-induced signaling in the tumor microenvironment in mice. In murine breast and colon carcinoma models, M7824 decreased tumor burden and increased overall survival as compared to targeting TGFbeta alone. M7824 treatment promoted CD8+ T cell and NK cell activation, and both of these immune populations were required for optimal M7824-mediated tumor control. M7824 was superior to TGFbeta- or alphaPD-L1-targeted therapies when in combination with a therapeutic cancer vaccine. These findings demonstrate the value of using M7824 to simultaneously target TGFbeta and PD-L1/PD-1 immunosuppressive pathways to promote anti-tumor responses and efficacy. The studies also support the potential clinical use of M7824 as a monotherapy or in combination with other immunotherapies, such as therapeutic cancer vaccines, including for patients who have progressed on alphaPD-L1/alphaPD-1 checkpoint blockade therapies.

Author Info: (1) Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. (2) Laboratory of Tumor Imm

Author Info: (1) Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. (2) Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. (3) Collaborative Protein Technology Resource (CPTR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. (4) Collaborative Protein Technology Resource (CPTR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. (5) Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. (6) Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.