Weiss and Puca et al. generated antibody-cytokine fusions (immunocytokines), combining L19 antibody (targeting extra-domain B of fibronectin) with IL-2, TNF, or IL-12 and demonstrated their antitumor activity. Intravenous L19-mIL12 and L19-mTNF suppressed tumor growth and prolonged survival in immunocompetent orthotopic glioma tumor models, but not in immunocompromised mice. L19-mIL12 increased proinflammatory cytokines and immune cell activation and infiltration into tumors. L19-mTNF showed direct cytotoxicity on tumor cells and increased immune cell infiltration. In 3 recurrent glioma patients, L19-hTNF was safe with indications of activity.

Contributed by Shishir Pant

ABSTRACT: Glioblastoma is a poorly immunogenic cancer, and the successes with recent immunotherapies in extracranial malignancies have, so far, not been translated to this devastating disease. Therefore, there is an urgent need for new strategies to convert the immunologically cold glioma microenvironment into a hot one to enable effective antitumor immunity. Using the L19 antibody, which is specific to a tumor-associated epitope of extracellular fibronectin, we developed antibody-cytokine fusions-immunocytokines-with interleukin-2 (IL2), IL12, or tumor necrosis factor (TNF). We showed that L19 accumulated in the tumor microenvironment of two orthotopic immunocompetent mouse glioma models. Furthermore, intravenous administration of L19-mIL12 or L19-mTNF cured a proportion of tumor-bearing mice, whereas L19-IL2 did not. This therapeutic activity was abolished in RAG(-/-) mice or upon depletion of CD4 or CD8 T cells, suggesting adaptive immunity. Mechanistically, both immunocytokines promoted tumor-infiltrating lymphocytes and increased the amounts of proinflammatory cytokines within the tumor microenvironment. In addition, L19-mTNF induced tumor necrosis. Systemic administration of the fully human L19-TNF fusion protein to patients with glioblastoma (NCT03779230) was safe, decreased regional blood perfusion within the tumor, and was associated with increasing tumor necrosis and an increase in tumor-infiltrating CD4 and CD8 T cells. The extensive preclinical characterization and subsequent clinical translation provide a robust basis for future studies with immunocytokines to treat malignant brain tumors.

Author Info: (1) Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland. (2) Department of Chemistry and Applied Biosci

Author Info: (1) Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland. (2) Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zrich), Vladimir-Prelog-Weg 4, CH-8093 Zrich, Switzerland. (3) Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland. (4) Philochem AG, CH-8112 Otelfingen, Switzerland. (5) Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland. (6) Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland. (7) Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland. (8) Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zrich), Vladimir-Prelog-Weg 4, CH-8093 Zrich, Switzerland. patrick.roth@usz.ch neri@pharma.ethz.ch. (9) Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland. patrick.roth@usz.ch neri@pharma.ethz.ch.